Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Nat Genet ; 55(9): 1440-1447, 2023 09.
Article in English | MEDLINE | ID: mdl-37537257

ABSTRACT

The incidence of keratinocyte cancer (basal cell and squamous cell carcinomas of the skin) is 17-fold lower in Singapore than the UK1-3, despite Singapore receiving 2-3 times more ultraviolet (UV) radiation4,5. Aging skin contains somatic mutant clones from which such cancers develop6,7. We hypothesized that differences in keratinocyte cancer incidence may be reflected in the normal skin mutational landscape. Here we show that, compared to Singapore, aging facial skin from populations in the UK has a fourfold greater mutational burden, a predominant UV mutational signature, increased copy number aberrations and increased mutant TP53 selection. These features are shared by keratinocyte cancers from high-incidence and low-incidence populations8-13. In Singaporean skin, most mutations result from cell-intrinsic processes; mutant NOTCH1 and NOTCH2 are more strongly selected than in the UK. Aging skin in a high-incidence country has multiple features convergent with cancer that are not found in a low-risk country. These differences may reflect germline variation in UV-protective genes.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Humans , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/genetics , Keratinocytes , Ultraviolet Rays/adverse effects , Mutation
3.
Nat Genet ; 55(2): 232-245, 2023 02.
Article in English | MEDLINE | ID: mdl-36658434

ABSTRACT

NOTCH1 mutant clones occupy the majority of normal human esophagus by middle age but are comparatively rare in esophageal cancers, suggesting NOTCH1 mutations drive clonal expansion but impede carcinogenesis. Here we test this hypothesis. Sequencing NOTCH1 mutant clones in aging human esophagus reveals frequent biallelic mutations that block NOTCH1 signaling. In mouse esophagus, heterozygous Notch1 mutation confers a competitive advantage over wild-type cells, an effect enhanced by loss of the second allele. Widespread Notch1 loss alters transcription but has minimal effects on the epithelial structure and cell dynamics. In a carcinogenesis model, Notch1 mutations were less prevalent in tumors than normal epithelium. Deletion of Notch1 reduced tumor growth, an effect recapitulated by anti-NOTCH1 antibody treatment. Notch1 null tumors showed reduced proliferation. We conclude that Notch1 mutations in normal epithelium are beneficial as wild-type Notch1 favors tumor expansion. NOTCH1 blockade may have therapeutic potential in preventing esophageal squamous cancer.


Subject(s)
Esophageal Neoplasms , Animals , Humans , Mice , Middle Aged , Carcinogenesis/pathology , Epithelium/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Mutation , Receptor, Notch1/genetics
4.
Cancer Discov ; 12(7): 1642-1655, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35397477

ABSTRACT

Epithelial stem cells accumulate mutations throughout life. Some of these mutants increase competitive fitness and may form clones that colonize the stem cell niche and persist to acquire further genome alterations. After a transient expansion, mutant stem cells must revert to homeostatic behavior so normal tissue architecture is maintained. Some positively selected mutants may promote cancer development, whereas others inhibit carcinogenesis. Factors that shape the mutational landscape include wild-type and mutant stem cell dynamics, competition for the niche, and environmental exposures. Understanding these processes may give new insight into the basis of cancer risk and opportunities for cancer prevention. SIGNIFICANCE: Recent advances in sequencing have found somatic mutations in all epithelial tissues studied to date. Here we review how the mutational landscape of normal epithelia is shaped by clonal competition within the stem cell niche combined with environmental exposures. Some of the selected mutant genes are oncogenic, whereas others may be inhibitory of transformation. Discoveries in this area leave many open questions, such as the definition of cancer driver genes, the mechanisms by which tissues constrain a high proportion of oncogenic mutant cells, and whether clonal fitness can be modulated to decrease cancer risk.


Subject(s)
Carcinogenesis , Neoplasms , Carcinogenesis/genetics , Clone Cells , Epithelium , Humans , Mutation , Neoplasms/genetics
5.
Cancer Discov ; 11(2): 340-361, 2021 02.
Article in English | MEDLINE | ID: mdl-33087317

ABSTRACT

Skin cancer risk varies substantially across the body, yet how this relates to the mutations found in normal skin is unknown. Here we mapped mutant clones in skin from high- and low-risk sites. The density of mutations varied by location. The prevalence of NOTCH1 and FAT1 mutations in forearm, trunk, and leg skin was similar to that in keratinocyte cancers. Most mutations were caused by ultraviolet light, but mutational signature analysis suggested differences in DNA-repair processes between sites. Eleven mutant genes were under positive selection, with TP53 preferentially selected in the head and FAT1 in the leg. Fine-scale mapping revealed 10% of clones had copy-number alterations. Analysis of hair follicles showed mutations in the upper follicle resembled adjacent skin, but the lower follicle was sparsely mutated. Normal skin is a dense patchwork of mutant clones arising from competitive selection that varies by location. SIGNIFICANCE: Mapping mutant clones across the body reveals normal skin is a dense patchwork of mutant cells. The variation in cancer risk between sites substantially exceeds that in mutant clone density. More generally, mutant genes cannot be assigned as cancer drivers until their prevalence in normal tissue is known.See related commentary by De Dominici and DeGregori, p. 227.This article is highlighted in the In This Issue feature, p. 211.


Subject(s)
Carcinoma, Basal Cell/genetics , Carcinoma, Squamous Cell/genetics , Skin Neoplasms/genetics , Adult , Aged , Cadherins/genetics , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Clone Cells , Female , Forearm , Humans , Leg , Male , Middle Aged , Mutation , Receptor, Notch1/genetics , Skin Neoplasms/pathology , Thorax
6.
Nat Genet ; 52(6): 604-614, 2020 06.
Article in English | MEDLINE | ID: mdl-32424351

ABSTRACT

During aging, progenitor cells acquire mutations, which may generate clones that colonize the surrounding tissue. By middle age, normal human tissues, including the esophageal epithelium (EE), become a patchwork of mutant clones. Despite their relevance for understanding aging and cancer, the processes that underpin mutational selection in normal tissues remain poorly understood. Here, we investigated this issue in the esophageal epithelium of mutagen-treated mice. Deep sequencing identified numerous mutant clones with multiple genes under positive selection, including Notch1, Notch2 and Trp53, which are also selected in human esophageal epithelium. Transgenic lineage tracing revealed strong clonal competition that evolved over time. Clone dynamics were consistent with a simple model in which the proliferative advantage conferred by positively selected mutations depends on the nature of the neighboring cells. When clones with similar competitive fitness collide, mutant cell fate reverts towards homeostasis, a constraint that explains how selection operates in normal-appearing epithelium.


Subject(s)
Esophagus/cytology , Mutation , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Animals , Cell Lineage , Diethylnitrosamine/toxicity , Epithelium/drug effects , Epithelium/pathology , Epithelium/physiology , Esophagus/physiology , Female , High-Throughput Nucleotide Sequencing , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Reproducibility of Results , Tumor Suppressor Protein p53/genetics
7.
Science ; 362(6417): 911-917, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30337457

ABSTRACT

The extent to which cells in normal tissues accumulate mutations throughout life is poorly understood. Some mutant cells expand into clones that can be detected by genome sequencing. We mapped mutant clones in normal esophageal epithelium from nine donors (age range, 20 to 75 years). Somatic mutations accumulated with age and were caused mainly by intrinsic mutational processes. We found strong positive selection of clones carrying mutations in 14 cancer genes, with tens to hundreds of clones per square centimeter. In middle-aged and elderly donors, clones with cancer-associated mutations covered much of the epithelium, with NOTCH1 and TP53 mutations affecting 12 to 80% and 2 to 37% of cells, respectively. Unexpectedly, the prevalence of NOTCH1 mutations in normal esophagus was several times higher than in esophageal cancers. These findings have implications for our understanding of cancer and aging.


Subject(s)
Aging/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagus/pathology , Selection, Genetic , Adult , Aged , Clone Cells/pathology , Female , Humans , Male , Middle Aged , Mutation , Receptor, Notch1/genetics , Tumor Suppressor Protein p53/genetics , Young Adult
8.
PLoS One ; 6(6): e20767, 2011.
Article in English | MEDLINE | ID: mdl-21698231

ABSTRACT

Here we investigate the effects of expressing an activated mutant of Notch (ICD-E) in an inducible transgenic mouse model. Hepatic expression of ICD-E in adult animals has no detectable phenotype, but simultaneous induction of ICD-E in both the liver and small intestine results in hepatic steatosis, lipogranuloma formation and mild insulin resistance within 96 hours. This supports work that suggests that fatty liver disease may result from disruption of the gut-liver axis. In the intestine, ICD-E expression is known to produce a transient change in the proportion of goblet cells followed by shedding of the recombinant epithelium. We report additional intestinal transcriptional changes following ICD-E expression, finding significant transcriptional down-regulation of rpL29 (ribosomal protein L29), which is implicated in the regulation of intestinal flora. These results provide further evidence of a gut-liver axis in the development of fatty liver disease and insulin resistance and validate a new model for future studies of hepatic steatosis.


Subject(s)
Fatty Liver/metabolism , Insulin Resistance , Intestinal Mucosa/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Transcription, Genetic
9.
Stem Cells ; 26(5): 1174-85, 2008 May.
Article in English | MEDLINE | ID: mdl-18292213

ABSTRACT

Activation of Hox gene clusters is an early event in embryonic development since individual members play important roles in patterning of the body axis. Their functions require precise control of spatiotemporal expression to provide positional information for the cells of the developing embryo, and the manner by which this control is achieved has generated considerable interest. The situation is different in pluripotent cells, where HOX genes are not expressed but are held in potentio as bivalent chromatin domains, which are resolved upon differentiation to permit HOX cluster activation. In this study we have used differentiation of the pluripotent embryonal carcinoma cell line NTera2SP12 and the human embryonic stem cell line H9 to examine epigenetic changes that accompany activation of the HOXA cluster and show that specific genomic loci are marked by lysine methylation of histone H3 (H3K4 tri- and dimethyl, H3K9 trimethyl) and acetylation of histone H4 even in the undifferentiated cells. The precise locations of such modified histones may be involved in controlling the colinear expression of genes from the cluster.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Acetylation , Animals , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Line , Genome, Human/genetics , Histones/metabolism , Homeodomain Proteins/metabolism , Humans , Lysine/metabolism , Methylation , Mice
10.
Genome Res ; 17(6): 691-707, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17567990

ABSTRACT

We generated high-resolution maps of histone H3 lysine 9/14 acetylation (H3ac), histone H4 lysine 5/8/12/16 acetylation (H4ac), and histone H3 at lysine 4 mono-, di-, and trimethylation (H3K4me1, H3K4me2, H3K4me3, respectively) across the ENCODE regions. Studying each modification in five human cell lines including the ENCODE Consortium common cell lines GM06990 (lymphoblastoid) and HeLa-S3, as well as K562, HFL-1, and MOLT4, we identified clear patterns of histone modification profiles with respect to genomic features. H3K4me3, H3K4me2, and H3ac modifications are tightly associated with the transcriptional start sites (TSSs) of genes, while H3K4me1 and H4ac have more widespread distributions. TSSs reveal characteristic patterns of both types of modification present and the position relative to TSSs. These patterns differ between active and inactive genes and in particular the state of H3K4me3 and H3ac modifications is highly predictive of gene activity. Away from TSSs, modification sites are enriched in H3K4me1 and relatively depleted in H3K4me3 and H3ac. Comparison between cell lines identified differences in the histone modification profiles associated with transcriptional differences between the cell lines. These results provide an overview of the functional relationship among histone modifications and gene expression in human cells.


Subject(s)
Genome, Human/physiology , Histones/metabolism , Protein Processing, Post-Translational/physiology , Transcription, Genetic/physiology , HeLa Cells , Humans , K562 Cells
11.
Hum Genet ; 113(6): 473-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12942364

ABSTRACT

The highly heterogeneous epithelial mucins show considerable inter-individual variability attributable to allelic variations in their tandem repeat (TR) peptide domains. Most mucins are known to show variations in repeat number but variation in the sequence of the individual TRs is not as well characterised. Here, we have studied variation in the immunodominant PDTR motif in the TR domain of the membrane-associated "cancer" mucin MUC1 by using the Minisatellite Variant Repeat-Polymerase chain reaction (MVR-PCR) technique. We have fully or partially mapped two nucleotide changes that encode two amino-acid changes, PDTR to PESR, across the arrays of 149 alleles. A total of 103 different maps was obtained when these changes alone were considered and additional variations were also observed. Most maps showed blocks of PDTR repeats interspersed with PESR repeats, although these were possibly more irregular in the longer alleles that also tended to have more PESR repeats. This variability has potential functional consequences and possible implications for some individuals with respect to the efficacy of immune targetting and immune therapy.


Subject(s)
Genetic Variation , Mucin-1/genetics , Amino Acid Sequence , Base Sequence , Humans , Molecular Sequence Data , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...