Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
EMBO J ; 31(11): 2444-5, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22510881

ABSTRACT

In this issue of The EMBO Journal, Wilson et al (2012) elegantly discovered an important new axis for intestinal homeostasis and cancer, using an RNAi screen to enhance the RAS-induced multivulva (MUV) phenotype in Caenorhabditis elegans.


Subject(s)
Homeostasis/physiology , Intestines/physiology , Intracellular Signaling Peptides and Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Stem Cells/physiology , Tumor Suppressor Proteins/genetics , Vesicular Transport Proteins/physiology , Animals , Female , Humans , Male , Oxidoreductases
3.
Cell Rep ; 36(1): 109307, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233177

ABSTRACT

Competitive cell interactions play a crucial role in quality control during development and homeostasis. Here, we show that cancer cells use such interactions to actively eliminate wild-type intestine cells in enteroid monolayers and organoids. This apoptosis-dependent process boosts proliferation of intestinal cancer cells. The remaining wild-type population activates markers of primitive epithelia and transits to a fetal-like state. Prevention of this cell-state transition avoids elimination of wild-type cells and, importantly, limits the proliferation of cancer cells. Jun N-terminal kinase (JNK) signaling is activated in competing cells and is required for cell-state change and elimination of wild-type cells. Thus, cell competition drives growth of cancer cells by active out-competition of wild-type cells through forced cell death and cell-state change in a JNK-dependent manner.


Subject(s)
Carcinogenesis/pathology , Intestines/pathology , Organoids/pathology , Animals , Apoptosis , Carcinogenesis/metabolism , Cell Competition , Cell Line, Tumor , Fetus/pathology , MAP Kinase Signaling System , Male , Mice, Inbred C57BL , Mice, Transgenic , Organoids/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism
4.
Cell Rep ; 32(3): 107937, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32698002

ABSTRACT

Calorie restriction (CR) extends lifespan through several intracellular mechanisms, including increased DNA repair, leading to fewer DNA mutations that cause age-related pathologies. However, it remains unknown how CR acts on mutation retention at the tissue level. Here, we use Cre-mediated DNA recombination of the confetti reporter as proxy for neutral mutations and follow these mutations by intravital microscopy to identify how CR affects retention of mutations in the intestine. We find that CR leads to increased numbers of functional Lgr5+ stem cells that compete for niche occupancy, resulting in slower but stronger stem cell competition. Consequently, stem cells carrying neutral or Apc mutations encounter more wild-type competitors, thus increasing the chance that they get displaced from the niche to get lost over time. Thus, our data show that CR not only affects the acquisition of mutations but also leads to lower retention of mutations in the intestine.


Subject(s)
Caloric Restriction , Cell Competition , Intestines/cytology , Mutation/genetics , Stem Cells/cytology , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cell Count , Cell Lineage , Female , Intravital Microscopy , Male , Mice, Inbred C57BL
5.
Nat Cell Biol ; 21(11): 1413-1424, 2019 11.
Article in English | MEDLINE | ID: mdl-31685988

ABSTRACT

Tumours depend on altered rates of protein synthesis for growth and survival, which suggests that mechanisms controlling mRNA translation may be exploitable for therapy. Here, we show that loss of APC, which occurs almost universally in colorectal tumours, strongly enhances the dependence on the translation initiation factor eIF2B5. Depletion of eIF2B5 induces an integrated stress response and enhances translation of MYC via an internal ribosomal entry site. This perturbs cellular amino acid and nucleotide pools, strains energy resources and causes MYC-dependent apoptosis. eIF2B5 limits MYC expression and prevents apoptosis in APC-deficient murine and patient-derived organoids and in APC-deficient murine intestinal epithelia in vivo. Conversely, the high MYC levels present in APC-deficient cells induce phosphorylation of eIF2α via the kinases GCN2 and PKR. Pharmacological inhibition of GCN2 phenocopies eIF2B5 depletion and has therapeutic efficacy in tumour organoids, which demonstrates that a negative MYC-eIF2α feedback loop constitutes a targetable vulnerability of colorectal tumours.


Subject(s)
Colorectal Neoplasms/genetics , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2B/antagonists & inhibitors , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Feedback, Physiological , Female , HCT116 Cells , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays
6.
Oncogene ; 24(14): 2430-2, 2005 Mar 31.
Article in English | MEDLINE | ID: mdl-15735763

ABSTRACT

Recent studies using hypomorphic DNA methyltransferase 1 (DNMT1) alleles have suggested that strategies aiming to reduce DNA methylation may increase genomic instability and lymphomagenesis. Given our recent finding that loss of methyl-binding domain protein 2 (Mbd2) suppresses intestinal tumorigenesis, we have tested whether loss of Mbd2 increases lymphomagenesis by intercrossing Mbd2 deficient mice with p53 deficient and p53 heterozygous mice. Unlike DNMT1, loss of Mbd2 does not accelerate lymphomagenesis, arguing that MBD2 may represent a better potential therapeutic target than DNMT1.


Subject(s)
DNA-Binding Proteins/genetics , Lymphoma/pathology , Tumor Suppressor Protein p53/physiology , Animals , DNA Methylation , Lymphoma/genetics , Mice , Mice, Knockout , Tumor Suppressor Protein p53/genetics
7.
Oncogene ; 22(46): 7130-6, 2003 Oct 16.
Article in English | MEDLINE | ID: mdl-14562041

ABSTRACT

MBD4 was originally identified through its methyl binding domain, but has more recently been characterized as a thymine DNA glycosylase that interacts with the mismatch repair (MMR) protein MLH1. In vivo, MBD4 functions to reduce the mutability of methyl-CpG sites in the genome and mice deticient in MBD4 show increased intestinal tumorigenesis on an Apc(Min/+) background. As MLH1 and other MMR proteins have been functionally linked to apoptosis, we asked whether MBD4 also plays a role in mediating the apoptotic response within the murine small intestine. Mice deficient for MBD4 showed significantly reduced apoptotic responses 6 h following treatment with a range of cytotoxic agents including gamma-irradiation, cisplatin, temozolomide and 5-fluorouracil (5-FU). This leads to increased clonogenic survival in vivo in Mbd4(-/-) mice following exposure to either 5-FU or cisplatin. We next analysed the apoptotic response to 5-FU and temozolomide in doubly mutant Mbd4(-/-), Mlh1(-/-) mice but observed no additive decrease. The results imply that MBD4 and MLH1 lie in the same pathway and therefore that MMR-dependent apoptosis is mediated through MBD4. MBD4 deficiency also reduced the normal apoptotic response to gamma-irradiation, which we show is independent of Mlh1 status (at least in the murine small intestine), so suggesting that the reliance upon MBD4 may extend beyond MMR-mediated apoptosis. Our results establish a novel functional role for MBD4 in the cellular response to DNA damage and may have implications for its role in suppressing neoplasia.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , DNA Damage , DNA Repair/genetics , Dacarbazine/analogs & derivatives , Endodeoxyribonucleases/deficiency , Intestinal Mucosa/pathology , Intestine, Small/physiology , Animals , Cell Survival/drug effects , Cell Survival/radiation effects , Cisplatin/toxicity , Dacarbazine/toxicity , Endodeoxyribonucleases/genetics , Fluorouracil/toxicity , Intestinal Mucosa/drug effects , Intestine, Small/cytology , Intestine, Small/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Temozolomide
8.
Nat Med ; 21(1): 62-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25419707

ABSTRACT

Hereditary mixed polyposis syndrome (HMPS) is characterized by the development of mixed-morphology colorectal tumors and is caused by a 40-kb genetic duplication that results in aberrant epithelial expression of the gene encoding mesenchymal bone morphogenetic protein antagonist, GREM1. Here we use HMPS tissue and a mouse model of the disease to show that epithelial GREM1 disrupts homeostatic intestinal morphogen gradients, altering cell fate that is normally determined by position along the vertical epithelial axis. This promotes the persistence and/or reacquisition of stem cell properties in Lgr5-negative progenitor cells that have exited the stem cell niche. These cells form ectopic crypts, proliferate, accumulate somatic mutations and can initiate intestinal neoplasia, indicating that the crypt base stem cell is not the sole cell of origin of colorectal cancer. Furthermore, we show that epithelial expression of GREM1 also occurs in traditional serrated adenomas, sporadic premalignant lesions with a hitherto unknown pathogenesis, and these lesions can be considered the sporadic equivalents of HMPS polyps.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Stem Cell Niche/genetics , Animals , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mutation , Receptors, G-Protein-Coupled/genetics
SELECTION OF CITATIONS
SEARCH DETAIL