Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
EMBO Rep ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937629

ABSTRACT

The EMT-transcription factor ZEB1 is heterogeneously expressed in tumor cells and in cancer-associated fibroblasts (CAFs) in colorectal cancer (CRC). While ZEB1 in tumor cells regulates metastasis and therapy resistance, its role in CAFs is largely unknown. Combining fibroblast-specific Zeb1 deletion with immunocompetent mouse models of CRC, we observe that inflammation-driven tumorigenesis is accelerated, whereas invasion and metastasis in sporadic cancers are reduced. Single-cell transcriptomics, histological characterization, and in vitro modeling reveal a crucial role of ZEB1 in CAF polarization, promoting myofibroblastic features by restricting inflammatory activation. Zeb1 deficiency impairs collagen deposition and CAF barrier function but increases NFκB-mediated cytokine production, jointly promoting lymphocyte recruitment and immune checkpoint activation. Strikingly, the Zeb1-deficient CAF repertoire sensitizes to immune checkpoint inhibition, offering a therapeutic opportunity of targeting ZEB1 in CAFs and its usage as a prognostic biomarker. Collectively, we demonstrate that ZEB1-dependent plasticity of CAFs suppresses anti-tumor immunity and promotes metastasis.

2.
Gastroenterology ; 164(2): 214-227, 2023 02.
Article in English | MEDLINE | ID: mdl-36402192

ABSTRACT

BACKGROUND & AIMS: Epigenetic processes regulating gene expression contribute markedly to epithelial cell plasticity in colorectal carcinogenesis. The lysine methyltransferase SUV420H2 comprises an important regulator of epithelial plasticity and is primarily responsible for trimethylation of H4K20 (H4K20me3). Loss of H4K20me3 has been suggested as a hallmark of human cancer due to its interaction with DNMT1. However, the role of Suv4-20h2 in colorectal cancer is unknown. METHODS: We examined the alterations in histone modifications in patient-derived colorectal cancer organoids. Patient-derived colorectal cancer organoids and mouse intestinal organoids were genetically manipulated for functional studies in patient-derived xenograft and orthotopic transplantation. Gene expression profiling, micrococcal nuclease assay, and chromatin immunoprecipitation were performed to understand epigenetic regulation of chromatin states and gene expression in patient-derived and mouse intestinal organoids. RESULTS: We found that reduced H4K20me3 levels occurred predominantly in right-sided patient-derived colorectal cancer organoids, which were associated with increased chromatin accessibility. Re-compaction of chromatin by methylstat, a histone demethylase inhibitor, resulted in reduced growth selectively in subcutaneously grown tumors derived from right-sided cancers. Using mouse intestinal organoids, we confirmed that Suv4-20h2-mediated H4K20me3 is required for maintaining heterochromatin compaction and to prevent R-loop formation. Cross-species comparison of Suv4-20h2-depleted murine organoids with right-sided colorectal cancer organoids revealed a large overlap of gene signatures involved in chromatin silencing, DNA methylation, and stemness/Wnt signaling. CONCLUSIONS: Loss of Suv4-20h2-mediated H4K20me3 drives right-sided colorectal tumorigenesis through an epigenetically controlled mechanism of chromatin compaction. Our findings unravel a conceptually novel approach for subtype-specific therapy of this aggressive form of colorectal cancer.


Subject(s)
Colonic Neoplasms , Histone-Lysine N-Methyltransferase , Animals , Humans , Mice , Cell Transformation, Neoplastic/genetics , Chromatin/genetics , Colonic Neoplasms/genetics , Colorectal Neoplasms/genetics , Epigenesis, Genetic , Histones/metabolism , Heterografts , Histone-Lysine N-Methyltransferase/metabolism
3.
EMBO J ; 38(12)2019 06 17.
Article in English | MEDLINE | ID: mdl-31036555

ABSTRACT

Immunotherapy using chimeric antigen receptor (CAR)-engineered lymphocytes has shown impressive results in leukemia. However, for solid tumors such as colorectal cancer (CRC), new preclinical models are needed that allow to test CAR-mediated cytotoxicity in a tissue-like environment. Here, we developed a platform to study CAR cell cytotoxicity against 3-dimensional (3D) patient-derived colon organoids. Luciferase-based measurement served as a quantitative read-out for target cell viability. Additionally, we set up a confocal live imaging protocol to monitor effector cell recruitment and cytolytic activity at a single organoid level. As proof of principle, we demonstrated efficient targeting in diverse organoid models using CAR-engineered NK-92 cells directed toward a ubiquitous epithelial antigen (EPCAM). Tumor antigen-specific cytotoxicity was studied with CAR-NK-92 cells targeting organoids expressing EGFRvIII, a neoantigen found in several cancers. Finally, we tested a novel CAR strategy targeting FRIZZLED receptors that show increased expression in a subgroup of CRC tumors. Here, comparative killing assays with normal organoids failed to show tumor-specific activity. Taken together, we report a sensitive in vitro platform to evaluate CAR efficacy and tumor specificity in a personalized manner.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Cytotoxicity, Immunologic , Models, Biological , Organoids/pathology , Receptors, Chimeric Antigen/therapeutic use , Tissue Culture Techniques/methods , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/genetics , Genetic Therapy/methods , HEK293 Cells , Humans , Immunotherapy, Adoptive/methods , Primary Cell Culture/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Tissue Scaffolds/chemistry
4.
Nature ; 530(7590): 340-3, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26863187

ABSTRACT

Mammalian Wnt proteins are believed to act as short-range signals, yet have not been previously visualized in vivo. Self-renewal, proliferation and differentiation are coordinated along a putative Wnt gradient in the intestinal crypt. Wnt3 is produced specifically by Paneth cells. Here we have generated an epitope-tagged, functional Wnt3 knock-in allele. Wnt3 covers basolateral membranes of neighbouring stem cells. In intestinal organoids, Wnt3-transfer involves direct contact between Paneth cells and stem cells. Plasma membrane localization requires surface expression of Frizzled receptors, which in turn is regulated by the transmembrane E3 ligases Rnf43/Znrf3 and their antagonists Lgr4-5/R-spondin. By manipulating Wnt3 secretion and by arresting stem-cell proliferation, we demonstrate that Wnt3 mainly travels away from its source in a cell-bound manner through cell division, and not through diffusion. We conclude that stem-cell membranes constitute a reservoir for Wnt proteins, while Frizzled receptor turnover and 'plasma membrane dilution' through cell division shape the epithelial Wnt3 gradient.


Subject(s)
Cell Membrane/metabolism , Intestinal Mucosa/cytology , Stem Cell Niche , Stem Cells/cytology , Stem Cells/metabolism , Wnt Signaling Pathway , Wnt3 Protein/metabolism , Alleles , Animals , Cell Adhesion , Cell Division , Diffusion , Female , Frizzled Receptors/metabolism , Gene Knock-In Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Organoids/cytology , Organoids/metabolism , Paneth Cells/cytology , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Ubiquitin-Protein Ligases/metabolism , Wnt3 Protein/genetics
5.
Sci Adv ; 9(11): eadd8564, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36921054

ABSTRACT

Immune checkpoint inhibitors (ICIs) enhance anticancer immunity by releasing repressive signals into tumor microenvironments (TMEs). To be effective, ICIs require preexisting immunologically "hot" niches for tumor antigen presentation and lymphocyte recruitment. How the mutational landscape of cancer cells shapes these immunological niches remains poorly defined. We found in human and murine colorectal cancer (CRC) models that the superior antitumor immune response of mismatch repair (MMR)-deficient CRC required tumor cell-intrinsic activation of cGAS-STING signaling triggered by genomic instability. Subsequently, we synthetically enforced STING signaling in CRC cells with intact MMR signaling using constitutively active STING variants. Even in MMR-proficient CRC, genetically encoded gain-of-function STING was sufficient to induce cancer cell-intrinsic interferon signaling, local activation of antigen-presenting cells, recruitment of effector lymphocytes, and sensitization of previously "cold" TMEs to ICI therapy in vivo. Thus, our results introduce a rational strategy for modulating cancer cell-intrinsic programs via engineered STING enforcement to sensitize resistant tumors to ICI responsiveness.


Subject(s)
Colonic Neoplasms , Signal Transduction , Humans , Animals , Mice , Antigen Presentation , Antigen-Presenting Cells , Genomic Instability , Tumor Microenvironment
6.
Cancer Discov ; 13(10): 2192-2211, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37489084

ABSTRACT

In colorectal cancers, the tumor microenvironment plays a key role in prognosis and therapy efficacy. Patient-derived tumor organoids (PDTO) show enormous potential for preclinical testing; however, cultured tumor cells lose important characteristics, including the consensus molecular subtypes (CMS). To better reflect the cellular heterogeneity, we established the colorectal cancer organoid-stroma biobank of matched PDTOs and cancer-associated fibroblasts (CAF) from 30 patients. Context-specific phenotyping showed that xenotransplantation or coculture with CAFs improves the transcriptomic fidelity and instructs subtype-specific stromal gene expression. Furthermore, functional profiling in coculture exposed CMS4-specific therapeutic resistance to gefitinib and SN-38 and prognostic expression signatures. Chemogenomic library screening identified patient- and therapy-dependent mechanisms of stromal resistance including MET as a common target. Our results demonstrate that colorectal cancer phenotypes are encrypted in the cancer epithelium in a plastic fashion that strongly depends on the context. Consequently, CAFs are essential for a faithful representation of molecular subtypes and therapy responses ex vivo. SIGNIFICANCE: Systematic characterization of the organoid-stroma biobank provides a resource for context dependency in colorectal cancer. We demonstrate a colorectal cancer subtype memory of PDTOs that is independent of specific driver mutations. Our data underscore the importance of functional profiling in cocultures for improved preclinical testing and identification of stromal resistance mechanisms. This article is featured in Selected Articles from This Issue, p. 2109.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Humans , Biological Specimen Banks , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Tumor Cells, Cultured , Cancer-Associated Fibroblasts/metabolism , Organoids/pathology , Tumor Microenvironment/genetics
7.
Cancer Cell ; 40(2): 168-184.e13, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35120600

ABSTRACT

Standard cancer therapy targets tumor cells without considering possible damage on the tumor microenvironment that could impair therapy response. In rectal cancer patients we find that inflammatory cancer-associated fibroblasts (iCAFs) are associated with poor chemoradiotherapy response. Employing a murine rectal cancer model or patient-derived tumor organoids and primary stroma cells, we show that, upon irradiation, interleukin-1α (IL-1α) not only polarizes cancer-associated fibroblasts toward the inflammatory phenotype but also triggers oxidative DNA damage, thereby predisposing iCAFs to p53-mediated therapy-induced senescence, which in turn results in chemoradiotherapy resistance and disease progression. Consistently, IL-1 inhibition, prevention of iCAFs senescence, or senolytic therapy sensitizes mice to irradiation, while lower IL-1 receptor antagonist serum levels in rectal patients correlate with poor prognosis. Collectively, we unravel a critical role for iCAFs in rectal cancer therapy resistance and identify IL-1 signaling as an attractive target for stroma-repolarization and prevention of cancer-associated fibroblasts senescence.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Drug Resistance, Neoplasm , Rectal Neoplasms/metabolism , Tumor Microenvironment , Animals , Biomarkers , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cytokines/genetics , Cytokines/metabolism , DNA Damage , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Heterografts , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Neoadjuvant Therapy , Prognosis , Rectal Neoplasms/drug therapy , Rectal Neoplasms/etiology , Rectal Neoplasms/pathology , Signal Transduction , Tumor Microenvironment/genetics
8.
Cancer Res ; 80(24): 5569-5582, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33055221

ABSTRACT

Tumor progression is recognized as a result of an evolving cross-talk between tumor cells and their surrounding nontransformed stroma. Although Wnt signaling has been intensively studied in colorectal cancer, it remains unclear whether activity in the tumor-associated stroma contributes to malignancy. To specifically interfere with stromal signals, we generated Wnt-independent tumor organoids that secrete the Wnt antagonist Sfrp1. Subcutaneous transplantation into immunocompetent as well as immunodeficient mice resulted in a strong reduction of tumor growth. Histologic and transcriptomic analyses revealed that Sfrp1 induced an epithelial-mesenchymal transition (EMT) phenotype in tumor cells without affecting tumor-intrinsic Wnt signaling, suggesting involvement of nonimmune stromal cells. Blockage of canonical signaling using Sfrp1, Dkk1, or fibroblast-specific genetic ablation of ß-catenin strongly decreased the number of cancer-associated myofibroblasts (myCAF). Wnt activity in CAFs was linked with distinct subtypes, where low and high levels induced an inflammatory-like CAF (iCAF) subtype or contractile myCAFs, respectively. Coculture of tumor organoids with iCAFs resulted in significant upregulation of EMT markers, while myCAFs reverted this phenotype. In summary, we show that tumor growth and malignancy are differentially regulated via distinct fibroblast subtypes under the influence of juxtacrine Wnt signals. SIGNIFICANCE: This study provides evidence for Wnt-induced functional diversity of colorectal cancer-associated fibroblasts, representing a non-cell autonomous mechanism for colon cancer progression. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/24/5569/F1.large.jpg.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Colorectal Neoplasms/metabolism , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Phenotype , Wnt Signaling Pathway/genetics , Wnt3 Protein/metabolism , Animals , Cell Survival/genetics , Coculture Techniques , Colorectal Neoplasms/pathology , Culture Media, Conditioned , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Organoids/metabolism , Organoids/transplantation , Transduction, Genetic , Wnt3 Protein/genetics
9.
Cell Stem Cell ; 26(5): 782-792.e7, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32348727

ABSTRACT

Colorectal cancer (CRC) is characterized by prominent genetic and phenotypic heterogeneity between patients. To facilitate high-throughput genetic testing and functional identification of tumor drivers, we developed a platform for pooled CRISPR-Cas9 screening in human colon organoids. Using transforming growth factor ß (TGF-ß) resistance as a paradigm to establish sensitivity and scalability in vitro, we identified optimal conditions and strict guide RNA (gRNA) requirements for screening in 3D organoids. We then screened a pan-cancer tumor suppressor gene (TSG) library in pre-malignant organoids with APC-/-;KRASG12D mutations, which were xenografted to study clonal advantages in context of a complex tumor microenvironment. We identified TGFBR2 as the most prevalent TSG, followed by known and previously uncharacterized mediators of CRC growth. gRNAs were validated in a secondary screen using unique molecular identifiers (UMIs) to adjust for clonal drift and to distinguish clone size and abundance. Together, these findings highlight a powerful organoid-based platform for pooled CRISPR-Cas9 screening for patient-specific functional genomics.


Subject(s)
CRISPR-Cas Systems , Organoids , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Colon , Genes, Tumor Suppressor , Humans
10.
J Exp Med ; 216(3): 704-720, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30792186

ABSTRACT

Constitutive Wnt activation upon loss of Adenoma polyposis coli (APC) acts as main driver of colorectal cancer (CRC). Targeting Wnt signaling has proven difficult because the pathway is crucial for homeostasis and stem cell renewal. To distinguish oncogenic from physiological Wnt activity, we have performed transcriptome and proteome profiling in isogenic human colon organoids. Culture in the presence or absence of exogenous ligand allowed us to discriminate receptor-mediated signaling from the effects of CRISPR/Cas9-induced APC loss. We could catalog two nonoverlapping molecular signatures that were stable at distinct levels of stimulation. Newly identified markers for normal stem/progenitor cells and adenomas were validated by immunohistochemistry and flow cytometry. We found that oncogenic Wnt signals are associated with good prognosis in tumors of the consensus molecular subtype 2 (CMS2). In contrast, receptor-mediated signaling was linked to CMS4 tumors and poor prognosis. Together, our data represent a valuable resource for biomarkers that allow more precise stratification of Wnt responses in CRC.


Subject(s)
Colon/pathology , Colon/physiology , Colonic Neoplasms/mortality , Organoids/physiology , Wnt Signaling Pathway/physiology , Adenoma/genetics , Adenoma/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Biomarkers, Tumor/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation , Gene Knockout Techniques , Humans , Kaplan-Meier Estimate , Organoids/pathology , Wnt Signaling Pathway/genetics
11.
Cell Mol Gastroenterol Hepatol ; 6(4): 477-493.e1, 2018.
Article in English | MEDLINE | ID: mdl-30364784

ABSTRACT

Background & Aims: Microvillus inclusion disease (MVID) is a congenital intestinal malabsorption disorder caused by defective apical vesicular transport. Existing cellular models do not fully recapitulate this heterogeneous pathology. The aim of this study was to characterize 3-dimensional intestinal organoids that continuously generate polarized absorptive cells as an accessible and relevant model to investigate MVID. Methods: Intestinal organoids from Munc18-2/Stxbp2-null mice that are deficient for apical vesicular transport were subjected to enterocyte-specific differentiation protocols. Lentiviral rescue experiments were performed using human MUNC18-2 variants. Apical trafficking and microvillus formation were characterized by confocal and transmission electron microscopy. Spinning disc time-lapse microscopy was used to document the lifecycle of microvillus inclusions. Results: Loss of Munc18-2/Stxbp2 recapitulated the pathologic features observed in patients with MUNC18-2 deficiency. The defects were fully restored by transgenic wild-type human MUNC18-2 protein, but not the patient variant (P477L). Importantly, we discovered that the MVID phenotype was correlated with the degree of enterocyte differentiation: secretory vesicles accumulated already in crypt progenitors, while differentiated enterocytes showed an apical tubulovesicular network and enlarged lysosomes. Upon prolonged enterocyte differentiation, cytoplasmic F-actin-positive foci were observed that further progressed into classic microvillus inclusions. Time-lapse microscopy showed their dynamic formation by intracellular maturation or invagination of the apical or basolateral plasma membrane. Conclusions: We show that prolonged enterocyte-specific differentiation is required to recapitulate the entire spectrum of MVID. Primary organoids can provide a powerful model for this heterogeneous pathology. Formation of microvillus inclusions from multiple membrane sources showed an unexpected dynamic of the enterocyte brush border.


Subject(s)
Cell Differentiation , Enterocytes/pathology , Intestines/pathology , Malabsorption Syndromes/metabolism , Microvilli/pathology , Mucolipidoses/metabolism , Munc18 Proteins/deficiency , Munc18 Proteins/metabolism , Organoids/metabolism , Actins/metabolism , Animals , Cell Nucleus/metabolism , Enterocytes/metabolism , Humans , Lysosomes/metabolism , Malabsorption Syndromes/pathology , Mice, Knockout , Microvilli/metabolism , Microvilli/ultrastructure , Mucolipidoses/pathology , Organoids/pathology , Organoids/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL