Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nature ; 537(7619): 239-243, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27525555

ABSTRACT

Neutrophils, eosinophils and 'classical' monocytes collectively account for about 70% of human blood leukocytes and are among the shortest-lived cells in the body. Precise regulation of the lifespan of these myeloid cells is critical to maintain protective immune responses and minimize the deleterious consequences of prolonged inflammation. However, how the lifespan of these cells is strictly controlled remains largely unknown. Here we identify a long non-coding RNA that we termed Morrbid, which tightly controls the survival of neutrophils, eosinophils and classical monocytes in response to pro-survival cytokines in mice. To control the lifespan of these cells, Morrbid regulates the transcription of the neighbouring pro-apoptotic gene, Bcl2l11 (also known as Bim), by promoting the enrichment of the PRC2 complex at the Bcl2l11 promoter to maintain this gene in a poised state. Notably, Morrbid regulates this process in cis, enabling allele-specific control of Bcl2l11 transcription. Thus, in these highly inflammatory cells, changes in Morrbid levels provide a locus-specific regulatory mechanism that allows rapid control of apoptosis in response to extracellular pro-survival signals. As MORRBID is present in humans and dysregulated in individuals with hypereosinophilic syndrome, this long non-coding RNA may represent a potential therapeutic target for inflammatory disorders characterized by aberrant short-lived myeloid cell lifespan.


Subject(s)
Bcl-2-Like Protein 11/genetics , Myeloid Cells/cytology , Myeloid Cells/metabolism , RNA, Long Noncoding/genetics , Alleles , Animals , Antigens, Ly/metabolism , Apoptosis , Bcl-2-Like Protein 11/biosynthesis , Cell Survival , Down-Regulation , Eosinophils/cytology , Eosinophils/metabolism , Female , Humans , Male , Mice , Monocytes/cytology , Monocytes/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Promoter Regions, Genetic
2.
Genes Dev ; 28(6): 652-64, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24637118

ABSTRACT

The mammalian intestinal epithelium has a unique organization in which crypts harboring stem cells produce progenitors and finally clonal populations of differentiated cells. Remarkably, the epithelium is replaced every 3-5 d throughout adult life. Disrupted maintenance of the intricate balance of proliferation and differentiation leads to loss of epithelial integrity or barrier function or to cancer. There is a tight correlation between the epigenetic status of genes and expression changes during differentiation; however, the mechanism of how changes in DNA methylation direct gene expression and the progression from stem cells to their differentiated descendants is unclear. Using conditional gene ablation of the maintenance methyltransferase Dnmt1, we demonstrate that reducing DNA methylation causes intestinal crypt expansion in vivo. Determination of the base-resolution DNA methylome in intestinal stem cells and their differentiated descendants shows that DNA methylation is dynamic at enhancers, which are often associated with genes important for both stem cell maintenance and differentiation. We establish that the loss of DNA methylation at intestinal stem cell gene enhancers causes inappropriate gene expression and delayed differentiation.


Subject(s)
Cell Differentiation , DNA Methylation , Intestine, Small/cytology , Stem Cells/cytology , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Mice
3.
Development ; 142(12): 2163-72, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26023099

ABSTRACT

The DNA methyltransferase Dnmt1 maintains DNA methylation patterns and genomic stability in several in vitro cell systems. Ablation of Dnmt1 in mouse embryos causes death at the post-gastrulation stage; however, the functions of Dnmt1 and DNA methylation in organogenesis remain unclear. Here, we report that Dnmt1 is crucial during perinatal intestinal development. Loss of Dnmt1 in intervillus progenitor cells causes global hypomethylation, DNA damage, premature differentiation, apoptosis and, consequently, loss of nascent villi. We further confirm the crucial role of Dnmt1 during crypt development using the in vitro organoid culture system, and illustrate a clear differential requirement for Dnmt1 in immature versus mature organoids. These results demonstrate an essential role for Dnmt1 in maintaining genomic stability during intestinal development and the establishment of intestinal crypts.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation/genetics , Genomic Instability/genetics , Intestinal Mucosa/embryology , Organogenesis/genetics , Stem Cells/cytology , Animals , Apoptosis/genetics , Cell Proliferation/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Damage/genetics , Gene Expression Regulation, Developmental , Intestinal Mucosa/cytology , Mice , Mice, Knockout , Molecular Sequence Data
4.
Cell Mol Life Sci ; 72(21): 4139-56, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26220502

ABSTRACT

The intestinal epithelium is an ideal model system for the study of normal and pathological differentiation processes. The mammalian intestinal epithelium is a single cell layer comprising proliferative crypts and differentiated villi. The crypts contain both proliferating and quiescent stem cell populations that self-renew and produce all the differentiated cell types, which are replaced every 3-5 days. The genetics of intestinal development, homeostasis, and disease are well defined, but less is known about the contribution of epigenetics in modulating these processes. Epigenetics refers to heritable phenotypic traits, including gene expression, which are independent of mutations in the DNA sequence. We have known for several decades that human colorectal cancers contain hypomethylated DNA, but the causes and consequences of this phenomenon are not fully understood. In contrast, tumor suppressor gene promoters are often hypermethylated in colorectal cancer, resulting in decreased expression of the associated gene. In this review, we describe the role that epigenetics plays in intestinal homeostasis and disease, with an emphasis on results from mouse models. We highlight the importance of producing and analyzing next-generation sequencing data detailing the epigenome from intestinal stem cell to differentiated intestinal villus cell.


Subject(s)
Colorectal Neoplasms/genetics , Epigenesis, Genetic , Intestinal Diseases/genetics , Intestinal Mucosa/physiology , Animals , Cell Differentiation , Chromatin/metabolism , Chromatin/ultrastructure , Colorectal Neoplasms/pathology , DNA Methylation , Histones/genetics , Histones/metabolism , Humans , Intestinal Diseases/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Mammals , Mice , Neoplasms, Experimental/genetics , Nucleosomes/genetics , Nucleosomes/metabolism , Receptors, Notch/metabolism , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Signaling Pathway
5.
Mol Microbiol ; 88(3): 486-500, 2013 May.
Article in English | MEDLINE | ID: mdl-23517529

ABSTRACT

Bacterial exopolysaccharide synthesis is a prevalent and indispensible activity in many biological processes, including surface adhesion and biofilm formation. In Caulobacter crescentus, surface attachment and subsequent biofilm growth depend on the ability to synthesize an adhesive polar polysaccharide known as the holdfast. In this work, we show that polar polysaccharide synthesis is a conserved phenomenon among Alphaproteobacterial species closely related to C. crescentus. Among them, mutagenesis of Asticcacaulis biprosthecum showed that disruption of the hfsH gene, which encodes a putative polysaccharide deacetylase, leads to accumulation of holdfast in the culture supernatant. Examination of the hfsH deletion mutant in C. crescentus revealed that this strain synthesizes holdfast; however, like the A. biprosthecum hfsH mutant, the holdfasts are shed into the medium and have decreased adhesiveness and cohesiveness. Site-directed mutagenesis at the predicted catalytic site of C. crescentus HfsH phenocopied the ΔhfsH mutant and abolished the esterase activity of HfsH. In contrast, overexpression of HfsH increased cell adherence without increasing holdfast synthesis. We conclude that the polysaccharide deacetylase activity of HfsH is required for the adhesive and cohesive properties of the holdfast, as well as for the anchoring of the holdfast to the cell envelope.


Subject(s)
Adhesins, Bacterial/metabolism , Amidohydrolases/metabolism , Bacterial Adhesion , Caulobacter crescentus/metabolism , Polysaccharides, Bacterial/metabolism , Amidohydrolases/genetics , Biofilms , Blotting, Western , Caulobacter crescentus/genetics , Caulobacter crescentus/growth & development , Chromosome Mapping , Computational Biology , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Multigene Family , Mutagenesis, Site-Directed , Plasmids/genetics , Sequence Deletion
6.
Cancer Prev Res (Phila) ; 9(7): 534-46, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26883721

ABSTRACT

Intestinal cancer is a heterogeneous disease driven by genetic mutations and epigenetic changes. Approximately 80% of sporadic colorectal cancers are initiated by mutation and inactivation of the adenomatous polyposis coli (APC) gene, which results in unrestrained intestinal epithelial growth and formation of adenomas. Aberrant DNA methylation promotes cancer progression by the inactivation of tumor suppressor genes via promoter methylation. In addition, global DNA hypomethylation is often seen before the formation of adenomas, suggesting that it contributes to neoplastic transformation. Previous studies employed mice with a hypomorphic mutation in DNA methyltransferase 1 (Dnmt1), which exhibited constitutive global DNA hypomethylation and decreased tumorigenesis in the Apc(Min/+) mouse model of intestinal cancer. However, the consequences of intestinal epithelial-specific acute hypomethylation during Apc(Min/+) tumor initiation have not been reported. Using temporally controlled intestinal epithelial-specific gene ablation, we show that total loss of Dnmt1 in the Apc(Min/+) mouse model of intestinal cancer causes accelerated adenoma initiation. Deletion of Dnmt1 precipitates an acute response characterized by hypomethylation of repetitive elements and genomic instability, which surprisingly is followed by remethylation with time. Two months post-Dnmt1 ablation, mice display increased macroadenoma load, consistent with a role for Dnmt1 and DNA methylation in maintaining genomic stability. These data suggest that DNA hypomethylation plays a previously unappreciated role in intestinal adenoma initiation. Cancer Prev Res; 9(7); 534-46. ©2016 AACRSee related article by Lee and Laird, p. 509.


Subject(s)
Adenoma/genetics , Carcinogenesis/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Intestinal Neoplasms/genetics , Adenoma/pathology , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , Genomic Instability/genetics , Intestinal Neoplasms/pathology , Mice
7.
Elife ; 52016 Jan 25.
Article in English | MEDLINE | ID: mdl-26808831

ABSTRACT

Dnmt1 is critical for immediate postnatal intestinal development, but is not required for the survival of the adult intestinal epithelium, the only rapidly dividing somatic tissue for which this has been shown. Acute Dnmt1 deletion elicits dramatic hypomethylation and genomic instability. Recovery of DNA methylation state and intestinal health is dependent on the de novo methyltransferase Dnmt3b. Ablation of both Dnmt1 and Dnmt3b in the intestinal epithelium is lethal, while deletion of either Dnmt1 or Dnmt3b has no effect on survival. These results demonstrate that Dnmt1 and Dnmt3b cooperate to maintain DNA methylation and genomic integrity in the intestinal epithelium.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/deficiency , DNA (Cytosine-5-)-Methyltransferases/metabolism , Intestinal Mucosa/enzymology , Intestinal Mucosa/physiology , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , Gene Deletion , Mice, Knockout , Survival Analysis , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL