Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 82(10): 1953-1968, 2022 05 16.
Article in English | MEDLINE | ID: mdl-35570706

ABSTRACT

Micrometastases of colorectal cancer can remain dormant for years prior to the formation of actively growing, clinically detectable lesions (i.e., colonization). A better understanding of this step in the metastatic cascade could help improve metastasis prevention and treatment. Here we analyzed liver specimens of patients with colorectal cancer and monitored real-time metastasis formation in mouse livers using intravital microscopy to reveal that micrometastatic lesions are devoid of cancer stem cells (CSC). However, lesions that grow into overt metastases demonstrated appearance of de novo CSCs through cellular plasticity at a multicellular stage. Clonal outgrowth of patient-derived colorectal cancer organoids phenocopied the cellular and transcriptomic changes observed during in vivo metastasis formation. First, formation of mature CSCs occurred at a multicellular stage and promoted growth. Conversely, failure of immature CSCs to generate more differentiated cells arrested growth, implying that cellular heterogeneity is required for continuous growth. Second, early-stage YAP activity was required for the survival of organoid-forming cells. However, subsequent attenuation of early-stage YAP activity was essential to allow for the formation of cell type heterogeneity, while persistent YAP signaling locked micro-organoids in a cellularly homogenous and growth-stalled state. Analysis of metastasis formation in mouse livers using single-cell RNA sequencing confirmed the transient presence of early-stage YAP activity, followed by emergence of CSC and non-CSC phenotypes, irrespective of the initial phenotype of the metastatic cell of origin. Thus, establishment of cellular heterogeneity after an initial YAP-controlled outgrowth phase marks the transition to continuously growing macrometastases. SIGNIFICANCE: Characterization of the cell type dynamics, composition, and transcriptome of early colorectal cancer liver metastases reveals that failure to establish cellular heterogeneity through YAP-controlled epithelial self-organization prohibits the outgrowth of micrometastases. See related commentary by LeBleu, p. 1870.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Animals , Colorectal Neoplasms/pathology , Humans , Liver Neoplasms/metabolism , Mice , Neoplasm Micrometastasis/pathology , Neoplastic Stem Cells/pathology
2.
STAR Protoc ; 1(3): 100126, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377020

ABSTRACT

Patient-derived organoids maintain functional and phenotypic characteristics of the original tissue such as cell-type diversity. Here, we provide protocols on how to label intestinal (cancer) stem cells by integrating the stem cell ASCL2 reporter (STAR) into human and mouse genomes via two different strategies: (1) lentiviral transduction or (2) transposon-based integration. Organoid technology, in combination with the user-friendly nature of STAR, will facilitate basic research in human and mouse adult stem cell biology. For complete details on the use and execution of this protocol, please refer to Oost et al. (2018).


Subject(s)
Organoids/metabolism , Staining and Labeling/methods , Adult Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Humans , Intestinal Mucosa/diagnostic imaging , Intestines/cytology , Mice , Organoids/growth & development , Stem Cells/metabolism
3.
Dev Cell ; 54(4): 435-446, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32841594

ABSTRACT

Damage to the intestinal stem cell niche can result from mechanical stress, infections, chronic inflammation or cytotoxic therapies. Progenitor cells can compensate for insults to the stem cell population through dedifferentiation. The microenvironment modulates this regenerative response by influencing the activity of signaling pathways, including Wnt, Notch, and YAP/TAZ. For instance, mesenchymal cells and immune cells become more abundant after damage and secrete signaling molecules that promote the regenerative process. Furthermore, regeneration is influenced by the nutritional state, microbiome, and extracellular matrix. Here, we review how all these components cooperate to restore epithelial homeostasis in the intestine after injury.


Subject(s)
Cell Dedifferentiation/genetics , Intestines/growth & development , Regeneration/genetics , Stem Cells/cytology , Acyltransferases , Cell Cycle Proteins/genetics , Cell Lineage/genetics , Cellular Microenvironment/genetics , Humans , Intestines/cytology , Receptors, Notch/genetics , Transcription Factors/genetics , Wnt Signaling Pathway/genetics
4.
Cell Stem Cell ; 26(4): 569-578.e7, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32169167

ABSTRACT

Colorectal cancer stem cells (CSCs) express Lgr5 and display extensive stem cell-like multipotency and self-renewal and are thought to seed metastatic disease. Here, we used a mouse model of colorectal cancer (CRC) and human tumor xenografts to investigate the cell of origin of metastases. We found that most disseminated CRC cells in circulation were Lgr5- and formed distant metastases in which Lgr5+ CSCs appeared. This plasticity occurred independently of stemness-inducing microenvironmental factors and was indispensable for outgrowth, but not establishment, of metastases. Together, these findings show that most colorectal cancer metastases are seeded by Lgr5- cells, which display intrinsic capacity to become CSCs in a niche-independent manner and can restore epithelial hierarchies in metastatic tumors.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Biomarkers, Tumor , Humans , Neoplastic Stem Cells , Receptors, G-Protein-Coupled
5.
Cell ; 180(2): 233-247.e21, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31978343

ABSTRACT

Wnt dependency and Lgr5 expression define multiple mammalian epithelial stem cell types. Under defined growth factor conditions, such adult stem cells (ASCs) grow as 3D organoids that recapitulate essential features of the pertinent epithelium. Here, we establish long-term expanding venom gland organoids from several snake species. The newly assembled transcriptome of the Cape coral snake reveals that organoids express high levels of toxin transcripts. Single-cell RNA sequencing of both organoids and primary tissue identifies distinct venom-expressing cell types as well as proliferative cells expressing homologs of known mammalian stem cell markers. A hard-wired regional heterogeneity in the expression of individual venom components is maintained in organoid cultures. Harvested venom peptides reflect crude venom composition and display biological activity. This study extends organoid technology to reptilian tissues and describes an experimentally tractable model system representing the snake venom gland.


Subject(s)
Cell Culture Techniques/methods , Organoids/growth & development , Snake Venoms/metabolism , Adult Stem Cells/metabolism , Animals , Coral Snakes/metabolism , Gene Expression Profiling/methods , Organoids/metabolism , Salivary Glands/metabolism , Snake Venoms/genetics , Snakes/genetics , Snakes/growth & development , Stem Cells/metabolism , Toxins, Biological/genetics , Transcriptome/genetics
6.
Cell Rep ; 22(6): 1600-1614, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29425513

ABSTRACT

Organoid technology provides the possibility of culturing patient-derived colon tissue and colorectal cancers (CRCs) while maintaining all functional and phenotypic characteristics. Labeling stem cells, especially in normal and benign tumor organoids of human colon, is challenging and therefore limits maximal exploitation of organoid libraries for human stem cell research. Here, we developed STAR (stem cell Ascl2 reporter), a minimal enhancer/promoter element that reports transcriptional activity of ASCL2, a master regulator of LGR5+ intestinal stem cells. Using lentiviral infection, STAR drives specific expression in stem cells of normal organoids and in multiple engineered and patient-derived CRC organoids of different genetic makeup. STAR reveals that differentiation hierarchies and the potential for cell fate plasticity are present at all stages of human CRC development. Organoid technology, in combination with the user-friendly nature of STAR, will facilitate basic research into human adult stem cell biology.


Subject(s)
Colorectal Neoplasms/pathology , Disease Models, Animal , Intestines , Organoids/pathology , Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors , Genes, Reporter , Heterografts , Humans , Intestines/cytology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...