Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
2.
J Exp Med ; 220(2)2023 02 06.
Article in English | MEDLINE | ID: mdl-36480166

ABSTRACT

IL-17A-producing γδ T cells in mice consist primarily of Vγ6+ tissue-resident cells and Vγ4+ circulating cells. How these γδ T cell subsets are regulated during homeostasis and cancer remains poorly understood. Using single-cell RNA sequencing and flow cytommetry, we show that lung Vγ4+ and Vγ6+ cells from tumor-free and tumor-bearing mice express contrasting cell surface molecules as well as distinct co-inhibitory molecules, which function to suppress their expansion. Vγ6+ cells express constitutively high levels of PD-1, whereas Vγ4+ cells upregulate TIM-3 in response to tumor-derived IL-1ß and IL-23. Inhibition of either PD-1 or TIM-3 in mammary tumor-bearing mice increased Vγ6+ and Vγ4+ cell numbers, respectively. We found that genetic deletion of γδ T cells elicits responsiveness to anti-PD-1 and anti-TIM-3 immunotherapy in a mammary tumor model that is refractory to T cell checkpoint inhibitors, indicating that IL-17A-producing γδ T cells instigate resistance to immunotherapy. Together, these data demonstrate how lung IL-17A-producing γδ T cell subsets are differentially controlled by PD-1 and TIM-3 in steady-state and cancer.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Interleukin-17 , Neoplasms , Programmed Cell Death 1 Receptor , T-Lymphocyte Subsets , Animals , Mice , Programmed Cell Death 1 Receptor/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism
3.
Cancers (Basel) ; 14(3)2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35159100

ABSTRACT

There is a growing body of evidence that cancer causes systemic changes. These influences are most evident in the bone marrow and the blood, particularly in the myeloid compartment. Here, we show that there is an increase in the number of bone marrow, circulating and splenic monocytes by using mouse models of breast cancer caused by the mammary epithelial expression of the polyoma middle T antigen. Cancer does not affect ratios of classical to non-classical populations of monocytes in the circulation nor does it affect their half-lives. Single cell RNA sequencing also indicates that cancer does not induce any new monocyte populations. Cancer does not change the monocytic progenitor number in the bone marrow, but the proliferation rate of monocytes is higher, thus providing an explanation for the expansion of the circulating numbers. Deep RNA sequencing of these monocytic populations reveals that cancer causes changes in the classical monocyte compartment, with changes evident in bone marrow monocytes and even more so in the blood, suggesting influences in both compartments, with the down-regulation of interferon type 1 signaling and antigen presentation being the most prominent of these. Consistent with this analysis, down-regulated genes are enriched with STAT1/STAT2 binding sites in their promoter, which are transcription factors required for type 1 interferon signaling. However, these transcriptome changes in mice did not replicate those found in patients with breast cancer. Consequently, this mouse model of breast cancer may be insufficient to study the systemic influences of human cancer.

4.
Life Sci Alliance ; 4(1)2021 01.
Article in English | MEDLINE | ID: mdl-33268347

ABSTRACT

The association of increased levels of tumour-infiltrating gamma-delta (γδ) T cells with favorable prognosis across many cancer types and their ability to recognize stress antigens in an MHC unrestricted manner has led to an increased interest in exploiting them for cancer immunotherapy. We performed single-cell RNA sequencing (scRNA-seq) of peripheral blood γδ T cells from healthy adult donors and from fresh tumour biopsies of breast cancer patients. We identified five γδ T cells subtypes in blood and three subtypes of γδ T cells in breast tumour. These subtypes differed in the expression of genes contributing to effector functions such as antigen presentation, cytotoxicity, and IL17A and IFNγ production. Compared with the blood γδ T cells, the breast tumour-infiltrating γδ T cells were more activated, expressed higher levels of cytotoxic genes, yet were immunosuppressed. One subtype in the breast tumour that was IFNγ-positive had no obvious similarity to any of the subtypes observed in the blood γδ T cell and was the only subtype associated with improved overall survival of breast cancer patients. Taken together, our study has identified markers of subtypes of human blood γδ T cells and uncovered a tumour-infiltrating γδ T cells subtype associated improved overall cancer survival.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/immunology , Intraepithelial Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , RNA-Seq/methods , Receptors, Antigen, T-Cell, gamma-delta/genetics , Single-Cell Analysis/methods , Adult , Base Sequence , Blood Donors , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Case-Control Studies , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Prognosis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
5.
iScience ; 23(3): 100914, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32151972

ABSTRACT

The power of single-cell RNA sequencing (scRNA-seq) stems from its ability to uncover cell type-dependent phenotypes, which rests on the accuracy of cell type identification. However, resolving cell types within and, thus, comparison of scRNA-seq data across conditions is challenging owing to technical factors such as sparsity, low number of cells, and batch effect. To address these challenges, we developed scID (Single Cell IDentification), which uses the Fisher's Linear Discriminant Analysis-like framework to identify transcriptionally related cell types between scRNA-seq datasets. We demonstrate the accuracy and performance of scID relative to existing methods on several published datasets. By increasing power to identify transcriptionally similar cell types across datasets with batch effect, scID enhances investigator's ability to integrate and uncover development-, disease-, and perturbation-associated changes in scRNA-seq data.

6.
J Transl Med ; 18(1): 45, 2020 01 30.
Article in English | MEDLINE | ID: mdl-32000794

ABSTRACT

The PTEN tumor suppressor is the second most commonly inactivated gene across cancer types. While it's role in PI3K/AKT and DNA damage pathways are clear, increasing evidences suggest that PTEN may also promote anti-tumor immunity. PTEN-deficient tumors are characterized by (i) reduced levels of cytotoxic T cells, helper T cells and NK cells, (ii) elevated pro-oncogenic inflammatory cytokines like CCL2 and (iii) increased levels of immunosuppressive cells such as MDSCs and Tregs. An intriguing possibility is that link between PTEN and anti-tumor immunity is mediated by the interferon signaling pathway. In this review, we summarize the evidences for the mechanistic link between PTEN deficiency and immunosuppressive tumor microenvironment and the interferon signaling pathway. We further discuss how the link between these pathways can be exploited for development of personalized immunotherapy for patients with PTEN deficient tumors.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Humans , Immunotherapy , Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases , Tumor Microenvironment
7.
Cell Cycle ; 19(1): 15-23, 2020 01.
Article in English | MEDLINE | ID: mdl-31760894

ABSTRACT

The DNA damage response (DDR) associated post-translational modifications recruit chromatin remodelers, signaling proteins such as 53BP1 and repair factors to chromatin flanking DNA double strand breaks (DSBs) to promote its repair. Although localization of both RNF168 ubiquitin ligase and SET8 methyltransferase at DSBs is essential for 53BP1's recruitment to DSBs, it is unclear if they do so via the same pathways. Here we report that RNF168 mediates SET8's recruitment to DSBs. Depletion of cellular pool of ubiquitin through proteasome inhibition abolished RNF168 and SET8's localization to DNA damage. Knockdown of RNF8 or RNF168 abolished SET8's recruitment to DNA damage. Moreover, RNF168 and SET8 form stable complexes in vivo. Based on these results we propose a model in which SET8, which despite being a pan-chromatin binding protein, can accumulate several folds at chromatin flanking DSBs through tethering to other proteins that specifically localize to chromatin regions with specific modifications.


Subject(s)
Chromatin/metabolism , DNA Damage , Histone-Lysine N-Methyltransferase/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , Humans , Models, Biological , Protein Binding , Protein Transport , Ubiquitin/metabolism
8.
Genes Dev ; 33(9-10): 498-510, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30842215

ABSTRACT

Developmental signal transduction pathways act diversely, with context-dependent roles across systems and disease types. Glioblastomas (GBMs), which are the poorest prognosis primary brain cancers, strongly resemble developmental systems, but these growth processes have not been exploited therapeutically, likely in part due to the extreme cellular and genetic heterogeneity observed in these tumors. The role of Wnt/ßcatenin signaling in GBM stem cell (GSC) renewal and fate decisions remains controversial. Here, we report context-specific actions of Wnt/ßcatenin signaling in directing cellular fate specification and renewal. A subset of primary GBM-derived stem cells requires Wnt proteins for self-renewal, and this subset specifically relies on Wnt/ßcatenin signaling for enhanced tumor burden in xenograft models. In an orthotopic Wnt reporter model, Wnthi GBM cells (which exhibit high levels of ßcatenin signaling) are a faster-cycling, highly self-renewing stem cell pool. In contrast, Wntlo cells (with low levels of signaling) are slower cycling and have decreased self-renewing potential. Dual inhibition of Wnt/ßcatenin and Notch signaling in GSCs that express high levels of the proneural transcription factor ASCL1 leads to robust neuronal differentiation and inhibits clonogenic potential. Our work identifies new contexts for Wnt modulation for targeting stem cell differentiation and self-renewal in GBM heterogeneity, which deserve further exploration therapeutically.


Subject(s)
Cell Differentiation/genetics , Neoplastic Stem Cells/cytology , Signal Transduction , Cell Line, Tumor , Cell Self Renewal/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/physiopathology , Humans , Receptors, Notch/genetics , Receptors, Notch/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
9.
Genome Biol ; 18(1): 213, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29115978

ABSTRACT

BACKGROUND: An important goal of cancer genomics is to identify systematically cancer-causing mutations. A common approach is to identify sites with high ratios of non-synonymous to synonymous mutations; however, if synonymous mutations are under purifying selection, this methodology leads to identification of false-positive mutations. Here, using synonymous somatic mutations (SSMs) identified in over 4000 tumours across 15 different cancer types, we sought to test this assumption by focusing on coding regions required for splicing. RESULTS: Exon flanks, which are enriched for sequences required for splicing fidelity, have ~ 17% lower SSM density compared to exonic cores, even after excluding canonical splice sites. While it is impossible to eliminate a mutation bias of unknown cause, multiple lines of evidence support a purifying selection model above a mutational bias explanation. The flank/core difference is not explained by skewed nucleotide content, replication timing, nucleosome occupancy or deficiency in mismatch repair. The depletion is not seen in tumour suppressors, consistent with their role in positive tumour selection, but is otherwise observed in cancer-associated and non-cancer genes, both essential and non-essential. Consistent with a role in splicing modulation, exonic splice enhancers have a lower SSM density before and after controlling for nucleotide composition; moreover, flanks at the 5' end of the exons have significantly lower SSM density than at the 3' end. CONCLUSIONS: These results suggest that the observable mutational spectrum of cancer genomes is not simply a product of various mutational processes and positive selection, but might also be shaped by negative selection.


Subject(s)
Genome, Human , Mutation/genetics , Neoplasms/genetics , RNA Splicing/genetics , Base Sequence , Enhancer Elements, Genetic/genetics , Exome/genetics , Exons/genetics , Humans , Nucleotide Motifs/genetics
10.
DNA Repair (Amst) ; 55: 64-75, 2017 07.
Article in English | MEDLINE | ID: mdl-28549257

ABSTRACT

Homology-directed repair (HDR) maintains genomic integrity by eliminating lesions such as DNA double-strand breaks (DSBs), interstrand crosslinks (ICLs) and stalled replication forks and thus a deficiency in HDR is associated with genomic instability and cancer predisposition. The mechanism of HDR is best understood and most rigorously characterized in yeast. The inactivation of the fungal radiation sensitive 52 (RAD52) gene, which has both recombination mediator and single-strand annealing (SSA) activities in vitro, leads to severe HDR defects in vivo. Confusingly, however, the inactivation of murine and chicken RAD52 genes resulted in mouse and chicken cells, respectively, that were largely aphenotypic. To clarify this issue, we have generated RAD52 knockout human cell lines. Human RAD52-null cells retain a significant level of SSA activity demonstrating perforce that additional SSA-like activities must exist in human cells. Moreover, we confirmed that the SSA activity associated with RAD52 is involved in, but not absolutely required for, most HDR subpathways. Specifically, a deficiency in RAD52 impaired the repair of DNA DSBs and intriguingly decreased the random integration of recombinant adeno-associated virus (rAAV). Finally, an analysis of pan-cancer genome data from The Cancer Genome Atlas (TCGA) revealed an association between aberrant levels of RAD52 expression and poor overall survival in multiple cancers. In toto, our work demonstrates that RAD52 contributes to the maintenance of genome stability and tumor suppression in human cells.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Neoplasms/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , DNA/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Genomic Instability , Humans , Neoplasms/genetics , Neoplasms/mortality , Rad52 DNA Repair and Recombination Protein/metabolism
11.
Nat Commun ; 6: 8036, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26292731

ABSTRACT

The generation of induced pluripotent stem cells (iPSC) from adult somatic cells is one of the most remarkable discoveries in recent decades. However, several works have reported evidence of genomic instability in iPSC, raising concerns on their biomedical use. The reasons behind the genomic instability observed in iPSC remain mostly unknown. Here we show that, similar to the phenomenon of oncogene-induced replication stress, the expression of reprogramming factors induces replication stress. Increasing the levels of the checkpoint kinase 1 (CHK1) reduces reprogramming-induced replication stress and increases the efficiency of iPSC generation. Similarly, nucleoside supplementation during reprogramming reduces the load of DNA damage and genomic rearrangements on iPSC. Our data reveal that lowering replication stress during reprogramming, genetically or chemically, provides a simple strategy to reduce genomic instability on mouse and human iPSC.


Subject(s)
Cell Proliferation/physiology , Cellular Reprogramming/physiology , Genomic Instability/physiology , Induced Pluripotent Stem Cells/physiology , Stress, Physiological/physiology , Animals , Cell Line , Checkpoint Kinase 1 , DNA/genetics , Fibroblasts/physiology , Gene Expression Regulation/physiology , Humans , Mice , Mice, Transgenic , Nucleic Acid Hybridization , Plasmids , Point Mutation , Protein Kinases/genetics , Protein Kinases/metabolism
12.
Cell Regen ; 3(1): 4, 2014.
Article in English | MEDLINE | ID: mdl-25408883

ABSTRACT

The conversion of somatic cells into pluripotent stem cells via overexpression of reprogramming factors involves epigenetic remodeling. DNA methylation at a significant proportion of CpG sites in induced pluripotent stem cells (iPSCs) differs from that of embryonic stem cells (ESCs). Whether different sets of reprogramming factors influence the type and extent of aberrant DNA methylation in iPSCs differently remains unknown. In order to help resolve this critical question, we generated human iPSCs from a common fibroblast cell source using either the Yamanaka factors (OCT4, SOX2, KLF4 and cMYC) or the Thomson factors (OCT4, SOX2, NANOG and LIN28), and determined their genome-wide DNA methylation profiles. In addition to shared DNA methylation aberrations present in all our iPSCs, we identified Yamanaka-iPSC (Y-iPSC)-specific and Thomson-iPSC (T-iPSC)-specific recurrent aberrations. Strikingly, not only were the genomic locations of the aberrations different but also their types: reprogramming with Yamanaka factors mainly resulted in failure to demethylate CpGs, whereas reprogramming with Thomson factors mainly resulted in failure to methylate CpGs. Differences in the level of transcripts encoding DNMT3b and TET3 between Y-iPSCs and T-iPSCs may contribute partially to the distinct types of aberrations. Finally, de novo aberrantly methylated genes in Y-iPSCs were enriched for NANOG targets that are also aberrantly methylated in some cancers. Our study thus reveals that the choice of reprogramming factors influences the amount, location, and class of DNA methylation aberrations in iPSCs. These findings may provide clues into how to produce human iPSCs with fewer DNA methylation abnormalities.

13.
EMBO Rep ; 15(11): 1163-74, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25252681

ABSTRACT

DNA double-strand breaks (DSBs) activate a signaling pathway known as the DNA damage response (DDR) which via protein-protein interactions and post-translational modifications recruit signaling proteins, such as 53BP1, to chromatin flanking the lesion. Depletion of the SET8 methyltransferase prevents accumulation of 53BP1 at DSBs; however, this phenotype has been attributed to the role of SET8 in generating H4K20 methylation across the genome, which is required for 53BP1 binding to chromatin, prior to DNA damage. Here, we report that SET8 acts directly at DSBs during the DNA damage response (DDR). SET8 accumulates at DSBs and is enzymatically active at DSBs. Depletion of SET8 just prior to the induction of DNA damage abrogates 53BP1's accumulation at DSBs, suggesting that SET8 acts during DDR. SET8's occupancy at DSBs is regulated by histone deacetylases (HDACs). Finally, SET8 is functionally required for efficient repair of DSBs specifically via the non-homologous end-joining pathway (NHEJ). Our findings reveal that SET8's active role during DDR at DSBs is required for 53BP1's accumulation.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Histone-Lysine N-Methyltransferase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Cell Line, Tumor , HEK293 Cells , Histone Deacetylases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Binding , Protein Transport , Tumor Suppressor p53-Binding Protein 1
14.
Stem Cell Reports ; 2(1): 44-51, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24511469

ABSTRACT

Somatic cells can be reprogrammed to induced pluripotent stem cells (iPSCs) using oncogenic transcription factors. However, this method leads to genetic aberrations in iPSCs via unknown mechanisms, which may limit their clinical use. Here, we demonstrate that the supplementation of growth media with antioxidants reduces the genome instability of cells transduced with the reprogramming factors. Antioxidant supplementation did not affect transgene expression level or silencing kinetics. Importantly, iPSCs made with antioxidants had significantly fewer de novo copy number variations, but not fewer coding point mutations, than iPSCs made without antioxidants. Our results suggest that the quality and safety of human iPSCs might be enhanced by using antioxidants in the growth media during the generation and maintenance of iPSCs.


Subject(s)
Antioxidants/pharmacology , Genomic Instability , Induced Pluripotent Stem Cells/drug effects , Cell Survival/drug effects , Cells, Cultured , Cellular Reprogramming/drug effects , DNA Damage/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Arch Otolaryngol Head Neck Surg ; 138(8): 732-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22911296

ABSTRACT

OBJECTIVE: To conduct high-throughput mutational analysis in 6 commonly used head and neck cancer cell lines. Comprehensive mutation analysis of primary head and neck squamous cell carcinoma (HNSCC) tumors has recently been reported, and mutations in the NOTCH receptors, TP53 and CDKN2A, were key findings. Established cell lines are valuable tools to study cancer in vitro. Similar high-throughput mutational analysis of head and neck cancer cell lines is necessary to confirm their mutational profile. DESIGN: DNA was extracted from American Type Culture Collection (ATCC) cell lines Cal27, Detroit562, FaDu, SCC4, SCC15, and SCC25. Cell line identity was confirmed by short tandem repeat (STR) analysis, and human papillomavirus (HPV) infection status was assessed by real-time polymerase chain reaction. A total of 535 cancer-associated genes were sequenced through a limited exome capture on the Illumina HiSeq system. SETTING: London Regional Cancer Program. RESULTS: The identity of the 6 cell lines was confirmed by STR analysis, and all lines tested negative for HPV infection. We achieved an average of 129-fold coverage with paired-end 100 base-pair reads. Sequencing revealed an average of 38 damaging mutations in each cell line (range, 30-45). The TP53 mutations, predicted to confer loss of function, were noted in all cell lines, and damaging CDKN2A mutations were found in all lines except SCC15. CONCLUSIONS: High-throughput sequencing of head and neck cancer cell lines revealed similar mutations to those observed in primary tumors. Thus, these lines reflect the tumor biology of HNSCC and can serve as valuable models to study HNSCC in vitro.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Mutational Analysis , Genes, p16 , Head and Neck Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Genotype , Head and Neck Neoplasms/virology , Humans , Microsatellite Repeats/genetics , Papillomaviridae/isolation & purification , Papillomavirus Infections/virology , Real-Time Polymerase Chain Reaction
16.
Stem Cells ; 30(3): 435-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22162363

ABSTRACT

Mutations in human induced pluripotent stem cells (iPSCs) pose a risk for their clinical use due to preferential reprogramming of mutated founder cell and selection of mutations during maintenance of iPSCs in cell culture. It is unknown, however, if mutations in iPSCs are due to stress associated with oncogene expression during reprogramming. We performed whole exome sequencing of human foreskin fibroblasts and their derived iPSCs at two different passages. We found that in vitro passaging contributed 7% to the iPSC coding point mutation load, and ultradeep amplicon sequencing revealed that 19% of the mutations preexist as rare mutations in the parental fibroblasts suggesting that the remaining 74% of the mutations were acquired during cellular reprogramming. Simulation suggests that the mutation intensity during reprogramming is ninefold higher than the background mutation rate in culture. Thus the factor induced reprogramming stress contributes to a significant proportion of the mutation load of iPSCs.


Subject(s)
Cell Dedifferentiation , Fibroblasts/physiology , Induced Pluripotent Stem Cells/cytology , Mutagenesis , Open Reading Frames/genetics , Cells, Cultured , DNA Mutational Analysis , Fibroblasts/metabolism , Genetic Vectors , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Octamer Transcription Factor-3/biosynthesis , Point Mutation , Proto-Oncogene Proteins c-myc/biosynthesis , Recombinant Proteins/biosynthesis , Retroviridae/genetics , SOXB1 Transcription Factors/biosynthesis
17.
Nature ; 471(7336): 58-62, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368824

ABSTRACT

The mechanisms underlying the low efficiency of reprogramming somatic cells into induced pluripotent stem (iPS) cells are poorly understood. There is a clear need to study whether the reprogramming process itself compromises genomic integrity and, through this, the efficiency of iPS cell establishment. Using a high-resolution single nucleotide polymorphism array, we compared copy number variations (CNVs) of different passages of human iPS cells with their fibroblast cell origins and with human embryonic stem (ES) cells. Here we show that significantly more CNVs are present in early-passage human iPS cells than intermediate passage human iPS cells, fibroblasts or human ES cells. Most CNVs are formed de novo and generate genetic mosaicism in early-passage human iPS cells. Most of these novel CNVs rendered the affected cells at a selective disadvantage. Remarkably, expansion of human iPS cells in culture selects rapidly against mutated cells, driving the lines towards a genetic state resembling human ES cells.


Subject(s)
Cellular Reprogramming/genetics , DNA Copy Number Variations/genetics , Induced Pluripotent Stem Cells/metabolism , Selection, Genetic , Cell Line , Chromosome Fragile Sites/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Haplotypes/genetics , Humans , In Situ Hybridization, Fluorescence , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/pathology , Mosaicism , Mutagenesis/genetics , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide/genetics , Selection, Genetic/genetics
18.
PLoS Genet ; 4(11): e1000250, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18989456

ABSTRACT

Coding sequence evolution was once thought to be the result of selection on optimal protein function alone. Selection can, however, also act at the RNA level, for example, to facilitate rapid translation or ensure correct splicing. Here, we ask whether the way DNA works also imposes constraints on coding sequence evolution. We identify nucleosome positioning as a likely candidate to set up such a DNA-level selective regime and use high-resolution microarray data in yeast to compare the evolution of coding sequence bound to or free from nucleosomes. Controlling for gene expression and intra-gene location, we find a nucleosome-free "linker" sequence to evolve on average 5-6% slower at synonymous sites. A reduced rate of evolution in linker is especially evident at the 5' end of genes, where the effect extends to non-synonymous substitution rates. This is consistent with regular nucleosome architecture in this region being important in the context of gene expression control. As predicted, codons likely to generate a sequence unfavourable to nucleosome formation are enriched in linker sequence. Amino acid content is likewise skewed as a function of nucleosome occupancy. We conclude that selection operating on DNA to maintain correct positioning of nucleosomes impacts codon choice, amino acid choice, and synonymous and non-synonymous rates of evolution in coding sequence. The results support the exclusion model for nucleosome positioning and provide an alternative interpretation for runs of rare codons. As the intimate association of histones and DNA is a universal characteristic of genic sequence in eukaryotes, selection on coding sequence composition imposed by nucleosome positioning should be phylogenetically widespread.


Subject(s)
Evolution, Molecular , Nucleosomes/genetics , Open Reading Frames , Saccharomyces cerevisiae/genetics , Gene Expression Regulation, Fungal , Genome, Fungal , Mutation , Selection, Genetic
19.
PLoS Biol ; 6(11): e277, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18998772

ABSTRACT

Previous studies in Saccharomyces cerevisiae have demonstrated that cryptic promoters within coding regions activate transcription in particular mutants. We have performed a comprehensive analysis of cryptic transcription in order to identify factors that normally repress cryptic promoters, to determine the amount of cryptic transcription genome-wide, and to study the potential for expression of genetic information by cryptic transcription. Our results show that a large number of factors that control chromatin structure and transcription are required to repress cryptic transcription from at least 1,000 locations across the S. cerevisiae genome. Two results suggest that some cryptic transcripts are translated. First, as expected, many cryptic transcripts contain an ATG and an open reading frame of at least 100 codons. Second, several cryptic transcripts are translated into proteins. Furthermore, a subset of cryptic transcripts tested is transiently induced in wild-type cells following a nutritional shift, suggesting a possible physiological role in response to a change in growth conditions. Taken together, our results demonstrate that, during normal growth, the global integrity of gene expression is maintained by a wide range of factors and suggest that, under altered genetic or physiological conditions, the expression of alternative genetic information may occur.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Fungal , Open Reading Frames/physiology , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Gene Expression Profiling , Genome, Fungal , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Open Reading Frames/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics
20.
Trends Genet ; 23(10): 480-4, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17822800

ABSTRACT

In diverse organisms, neighbouring genes in the genome tend to be positively coexpressed more than expected by chance. When the similarity of transcription regulation is controlled for, adjacent genes have much higher coexpression rates than unlinked genes, supporting a role for chromatin modelling. Consequently, many incidences of low-to-moderate level coexpression of linked genes might well be spurious rather than an indication of functional coordination. These results have implications for gene therapy and for understanding gene order evolution, suggesting that chromosomal proximity alone is adequate to achieve some level of coexpression.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae/genetics , Evolution, Molecular , Genome, Fungal , Nucleosomes/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...