Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Mol Cell ; 72(1): 37-47.e4, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30197296

ABSTRACT

Adenomatous polyposis coli (APC) and Axin are core components of the ß-catenin destruction complex. How APC's function is regulated and whether Wnt signaling influences the direct APC-Axin interaction to inhibit the ß-catenin destruction complex is not clear. Through a CRISPR screen of ß-catenin stability, we have identified ICAT, a polypeptide previously known to block ß-catenin-TCF interaction, as a natural inhibitor of APC. ICAT blocks ß-catenin-APC interaction and prevents ß-catenin-mediated APC-Axin interaction, enhancing stabilization of ß-catenin in cells harboring truncated APC or stimulated with Wnt, but not in cells deprived of a Wnt signal. Using ICAT as a tool to disengage ß-catenin-mediated APC-Axin interaction, we demonstrate that Wnt quickly inhibits the direct interaction between APC and Axin. Our study highlights an important scaffolding function of ß-catenin in the assembly of the destruction complex and suggests Wnt-inhibited APC-Axin interaction as a mechanism of Wnt-dependent inhibition of the destruction complex.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Interaction Domains and Motifs/genetics , beta Catenin/genetics , Adaptor Proteins, Signal Transducing , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Axin Protein/genetics , Humans , Protein Stability , Transcription Factor 7-Like 1 Protein/genetics , Wnt Signaling Pathway/genetics
2.
Mol Cell ; 63(4): 633-646, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27499295

ABSTRACT

The repair outcomes at site-specific DNA double-strand breaks (DSBs) generated by the RNA-guided DNA endonuclease Cas9 determine how gene function is altered. Despite the widespread adoption of CRISPR-Cas9 technology to induce DSBs for genome engineering, the resulting repair products have not been examined in depth. Here, the DNA repair profiles of 223 sites in the human genome demonstrate that the pattern of DNA repair following Cas9 cutting at each site is nonrandom and consistent across experimental replicates, cell lines, and reagent delivery methods. Furthermore, the repair outcomes are determined by the protospacer sequence rather than genomic context, indicating that DNA repair profiling in cell lines can be used to anticipate repair outcomes in primary cells. Chemical inhibition of DNA-PK enabled dissection of the DNA repair profiles into contributions from c-NHEJ and MMEJ. Finally, this work elucidates a strategy for using "error-prone" DNA-repair machinery to generate precise edits.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Cas Systems , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Endonucleases/metabolism , Gene Editing , Gene Expression Profiling/methods , Bacterial Proteins/genetics , CRISPR-Associated Protein 9 , Endonucleases/genetics , HCT116 Cells , HEK293 Cells , Humans , K562 Cells , RNA Interference , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Time Factors , Transfection
3.
Nat Methods ; 15(11): 941-946, 2018 11.
Article in English | MEDLINE | ID: mdl-30297964

ABSTRACT

CRISPR-Cas9 screening allows genome-wide interrogation of gene function. Currently, to achieve the high and uniform Cas9 expression desirable for screening, one needs to engineer stable and clonal Cas9-expressing cells-an approach that is not applicable in human primary cells. Guide Swap permits genome-scale pooled CRISPR-Cas9 screening in human primary cells by exploiting the unexpected finding that editing by lentivirally delivered, targeted guide RNAs (gRNAs) occurs efficiently when Cas9 is introduced in complex with nontargeting gRNA. We validated Guide Swap in depletion and enrichment screens in CD4+ T cells. Next, we implemented Guide Swap in a model of ex vivo hematopoiesis, and identified known and previously unknown regulators of CD34+ hematopoietic stem and progenitor cell (HSPC) expansion. We anticipate that this platform will be broadly applicable to other challenging cell types, and thus will enable discovery in previously inaccessible but biologically relevant human primary cell systems.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Gene Editing , Genome, Human , Hematopoietic Stem Cells/metabolism , RNA, Guide, Kinetoplastida/genetics , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans
4.
Proc Natl Acad Sci U S A ; 115(44): E10362-E10369, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30297426

ABSTRACT

Wnt/ß-catenin signaling plays pivotal roles in cell proliferation and tissue homeostasis by maintaining somatic stem cell functions. The mammalian target of rapamycin (mTOR) signaling functions as an integrative rheostat that orchestrates various cellular and metabolic activities that shape tissue homeostasis. Whether these two fundamental signaling pathways couple to exert physiological functions still remains mysterious. Using a genome-wide CRISPR-Cas9 screening, we discover that mTOR complex 1 (mTORC1) signaling suppresses canonical Wnt/ß-catenin signaling. Deficiency in tuberous sclerosis complex 1/2 (TSC1/2), core negative regulators of mTORC1 activity, represses Wnt/ß-catenin target gene expression, which can be rescued by RAD001. Mechanistically, mTORC1 signaling regulates the cell surface level of Wnt receptor Frizzled (FZD) in a Dishevelled (DVL)-dependent manner by influencing the association of DVL and clathrin AP-2 adaptor. Sustained mTORC1 activation impairs Wnt/ß-catenin signaling and causes loss of stemness in intestinal organoids ex vivo and primitive intestinal progenitors in vivo. Wnt/ß-catenin-dependent liver metabolic zonation gene expression program is also down-regulated by mTORC1 activation. Our study provides a paradigm that mTORC1 signaling cell autonomously regulates Wnt/ß-catenin pathway to influence stem cell maintenance.


Subject(s)
Frizzled Receptors/metabolism , Receptors, Wnt/metabolism , TOR Serine-Threonine Kinases/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Adaptor Protein Complex 2/metabolism , Animals , Cell Line , Dishevelled Proteins/metabolism , Down-Regulation/physiology , Gene Expression/physiology , HEK293 Cells , Humans , Mice
5.
Proc Natl Acad Sci U S A ; 115(2): E180-E189, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29269392

ABSTRACT

PARKIN, an E3 ligase mutated in familial Parkinson's disease, promotes mitophagy by ubiquitinating mitochondrial proteins for efficient engagement of the autophagy machinery. Specifically, PARKIN-synthesized ubiquitin chains represent targets for the PINK1 kinase generating phosphoS65-ubiquitin (pUb), which constitutes the mitophagy signal. Physiological regulation of PARKIN abundance, however, and the impact on pUb accumulation are poorly understood. Using cells designed to discover physiological regulators of PARKIN abundance, we performed a pooled genome-wide CRISPR/Cas9 knockout screen. Testing identified genes individually resulted in a list of 53 positive and negative regulators. A transcriptional repressor network including THAP11 was identified and negatively regulates endogenous PARKIN abundance. RNAseq analysis revealed the PARKIN-encoding locus as a prime THAP11 target, and THAP11 CRISPR knockout in multiple cell types enhanced pUb accumulation. Thus, our work demonstrates the critical role of PARKIN abundance, identifies regulating genes, and reveals a link between transcriptional repression and mitophagy, which is also apparent in human induced pluripotent stem cell-derived neurons, a disease-relevant cell type.


Subject(s)
CRISPR-Cas Systems , Gene Expression Regulation , Genome, Human/genetics , Mitophagy/genetics , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Cell Line, Tumor , Cells, Cultured , HCT116 Cells , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Infant, Newborn , Neurons/metabolism , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Repressor Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
6.
J Biol Chem ; 294(10): 3359-3366, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30647128

ABSTRACT

Bile acids are critical metabolites in the gastrointestinal tract and contribute to maintaining intestinal immune homeostasis through cross-talk with the gut microbiota. The conversion of bile acids by the gut microbiome is now recognized as a factor affecting both host metabolism and immune responses, but its physiological roles remain unclear. We conducted a screen for microbiome metabolites that would function as inflammasome activators and herein report the identification of 12-oxo-lithocholic acid (BAA485), a potential microbiome-derived bile acid metabolite. We demonstrate that the more potent analogue 11-oxo-12S-hydroxylithocholic acid methyl ester (BAA473) can induce secretion of interleukin-18 (IL-18) through activation of the inflammasome in both myeloid and intestinal epithelial cells. Using a genome-wide CRISPR screen with compound induced pyroptosis in THP-1 cells, we identified that inflammasome activation by BAA473 is pyrin-dependent (MEFV). To our knowledge, the bile acid analogues BAA485 and BAA473 are the first small molecule activators of the pyrin inflammasome. We surmise that pyrin inflammasome activation through microbiota-modified bile acid metabolites such as BAA473 and BAA485 plays a role in gut microbiota regulated intestinal immune response. The discovery of these two bioactive compounds may help to further unveil the importance of pyrin in gut homeostasis and autoimmune diseases.


Subject(s)
Bile Acids and Salts/immunology , Epithelial Cells/immunology , Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Inflammasomes/immunology , Intestinal Mucosa/immunology , Pyrin/immunology , Bile Acids and Salts/chemistry , Humans , Myeloid Cells/immunology , THP-1 Cells
7.
EMBO Rep ; 19(9)2018 09.
Article in English | MEDLINE | ID: mdl-30126924

ABSTRACT

Autophagy maintains cellular homeostasis by targeting damaged organelles, pathogens, or misfolded protein aggregates for lysosomal degradation. The autophagic process is initiated by the formation of autophagosomes, which can selectively enclose cargo via autophagy cargo receptors. A machinery of well-characterized autophagy-related proteins orchestrates the biogenesis of autophagosomes; however, the origin of the required membranes is incompletely understood. Here, we have applied sensitized pooled CRISPR screens and identify the uncharacterized transmembrane protein TMEM41B as a novel regulator of autophagy. In the absence of TMEM41B, autophagosome biogenesis is stalled, LC3 accumulates at WIPI2- and DFCP1-positive isolation membranes, and lysosomal flux of autophagy cargo receptors and intracellular bacteria is impaired. In addition to defective autophagy, TMEM41B knockout cells display significantly enlarged lipid droplets and reduced mobilization and ß-oxidation of fatty acids. Immunostaining and interaction proteomics data suggest that TMEM41B localizes to the endoplasmic reticulum (ER). Taken together, we propose that TMEM41B is a novel ER-localized regulator of autophagosome biogenesis and lipid mobilization.


Subject(s)
Autophagy/physiology , Lipid Mobilization/physiology , Membrane Proteins/genetics , Membrane Proteins/physiology , Autophagosomes/metabolism , Autophagy/genetics , Autophagy-Related Proteins/metabolism , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Endoplasmic Reticulum/metabolism , Fatty Acids/metabolism , Gene Knockout Techniques , HeLa Cells , Homeostasis , Humans , Lentivirus , Lipid Droplets/metabolism , Lipid Mobilization/genetics , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism
8.
Nature ; 488(7409): 106-10, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22820256

ABSTRACT

Medulloblastomas are the most common malignant brain tumours in children. Identifying and understanding the genetic events that drive these tumours is critical for the development of more effective diagnostic, prognostic and therapeutic strategies. Recently, our group and others described distinct molecular subtypes of medulloblastoma on the basis of transcriptional and copy number profiles. Here we use whole-exome hybrid capture and deep sequencing to identify somatic mutations across the coding regions of 92 primary medulloblastoma/normal pairs. Overall, medulloblastomas have low mutation rates consistent with other paediatric tumours, with a median of 0.35 non-silent mutations per megabase. We identified twelve genes mutated at statistically significant frequencies, including previously known mutated genes in medulloblastoma such as CTNNB1, PTCH1, MLL2, SMARCA4 and TP53. Recurrent somatic mutations were newly identified in an RNA helicase gene, DDX3X, often concurrent with CTNNB1 mutations, and in the nuclear co-repressor (N-CoR) complex genes GPS2, BCOR and LDB1. We show that mutant DDX3X potentiates transactivation of a TCF promoter and enhances cell viability in combination with mutant, but not wild-type, ß-catenin. Together, our study reveals the alteration of WNT, hedgehog, histone methyltransferase and now N-CoR pathways across medulloblastomas and within specific subtypes of this disease, and nominates the RNA helicase DDX3X as a component of pathogenic ß-catenin signalling in medulloblastoma.


Subject(s)
Cerebellar Neoplasms/genetics , Exome/genetics , Genome, Human/genetics , Medulloblastoma/genetics , Mutation/genetics , Cerebellar Neoplasms/classification , Child , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA Helicases/chemistry , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Hedgehog Proteins/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Medulloblastoma/classification , Models, Molecular , Neoplasm Proteins/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Patched Receptors , Patched-1 Receptor , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins/genetics , Receptors, Cell Surface/genetics , Repressor Proteins/genetics , Signal Transduction , TCF Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
9.
Genes Dev ; 24(10): 992-1009, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20413612

ABSTRACT

MicroRNAs (miRNAs) are small regulatory RNAs that derive from distinctive hairpin transcripts. To learn more about the miRNAs of mammals, we sequenced 60 million small RNAs from mouse brain, ovary, testes, embryonic stem cells, three embryonic stages, and whole newborns. Analysis of these sequences confirmed 398 annotated miRNA genes and identified 108 novel miRNA genes. More than 150 previously annotated miRNAs and hundreds of candidates failed to yield sequenced RNAs with miRNA-like features. Ectopically expressing these previously proposed miRNA hairpins also did not yield small RNAs, whereas ectopically expressing the confirmed and newly identified hairpins usually did yield small RNAs with the classical miRNA features, including dependence on the Drosha endonuclease for processing. These experiments, which suggest that previous estimates of conserved mammalian miRNAs were inflated, provide a substantially revised list of confidently identified murine miRNAs from which to infer the general features of mammalian miRNAs. Our analyses also revealed new aspects of miRNA biogenesis and modification, including tissue-specific strand preferences, sequential Dicer cleavage of a metazoan precursor miRNA (pre-miRNA), consequential 5' heterogeneity, newly identified instances of miRNA editing, and evidence for widespread pre-miRNA uridylation reminiscent of miRNA regulation by Lin28.


Subject(s)
Genes/genetics , Genome/genetics , MicroRNAs/genetics , Animals , Cell Line , Gene Expression Profiling , Humans , Inverted Repeat Sequences/genetics , Mice , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Ribonuclease III/metabolism
10.
J Biol Chem ; 291(29): 15256-66, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27231341

ABSTRACT

YAP signaling pathway plays critical roles in tissue homeostasis, and aberrant activation of YAP signaling has been implicated in cancers. To identify tractable targets of YAP pathway, we have performed a pathway-based pooled CRISPR screen and identified tankyrase and its associated E3 ligase RNF146 as positive regulators of YAP signaling. Genetic ablation or pharmacological inhibition of tankyrase prominently suppresses YAP activity and YAP target gene expression. Using a proteomic approach, we have identified angiomotin family proteins, which are known negative regulators of YAP signaling, as novel tankyrase substrates. Inhibition of tankyrase or depletion of RNF146 stabilizes angiomotins. Angiomotins physically interact with tankyrase through a highly conserved motif at their N terminus, and mutation of this motif leads to their stabilization. Tankyrase inhibitor-induced stabilization of angiomotins reduces YAP nuclear translocation and decreases downstream YAP signaling. We have further shown that knock-out of YAP sensitizes non-small cell lung cancer to EGFR inhibitor Erlotinib. Tankyrase inhibitor, but not porcupine inhibitor, which blocks Wnt secretion, enhances growth inhibitory activity of Erlotinib. This activity is mediated by YAP inhibition and not Wnt/ß-catenin inhibition. Our data suggest that tankyrase inhibition could serve as a novel strategy to suppress YAP signaling for combinatorial targeted therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Phosphoproteins/antagonists & inhibitors , Tankyrases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Angiomotins , Antineoplastic Agents/pharmacology , CRISPR-Cas Systems , Cell Line, Tumor , Down-Regulation , Erlotinib Hydrochloride/pharmacology , Gene Knockout Techniques , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microfilament Proteins , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Interaction Domains and Motifs , Protein Stability/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Tankyrases/chemistry , Tankyrases/genetics , Transcription Factors , Ubiquitin-Protein Ligases/metabolism , YAP-Signaling Proteins
11.
PLoS Genet ; 9(6): e1003272, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23785293

ABSTRACT

Oomycetes in the class Saprolegniomycetidae of the Eukaryotic kingdom Stramenopila have evolved as severe pathogens of amphibians, crustaceans, fish and insects, resulting in major losses in aquaculture and damage to aquatic ecosystems. We have sequenced the 63 Mb genome of the fresh water fish pathogen, Saprolegnia parasitica. Approximately 1/3 of the assembled genome exhibits loss of heterozygosity, indicating an efficient mechanism for revealing new variation. Comparison of S. parasitica with plant pathogenic oomycetes suggests that during evolution the host cellular environment has driven distinct patterns of gene expansion and loss in the genomes of plant and animal pathogens. S. parasitica possesses one of the largest repertoires of proteases (270) among eukaryotes that are deployed in waves at different points during infection as determined from RNA-Seq data. In contrast, despite being capable of living saprotrophically, parasitism has led to loss of inorganic nitrogen and sulfur assimilation pathways, strikingly similar to losses in obligate plant pathogenic oomycetes and fungi. The large gene families that are hallmarks of plant pathogenic oomycetes such as Phytophthora appear to be lacking in S. parasitica, including those encoding RXLR effectors, Crinkler's, and Necrosis Inducing-Like Proteins (NLP). S. parasitica also has a very large kinome of 543 kinases, 10% of which is induced upon infection. Moreover, S. parasitica encodes several genes typical of animals or animal-pathogens and lacking from other oomycetes, including disintegrins and galactose-binding lectins, whose expression and evolutionary origins implicate horizontal gene transfer in the evolution of animal pathogenesis in S. parasitica.


Subject(s)
Gene Transfer, Horizontal , Host-Parasite Interactions/genetics , Oomycetes/genetics , Saprolegnia/genetics , Virulence/genetics , Amino Acid Sequence , Animals , Base Sequence , Evolution, Molecular , Fishes/genetics , Fishes/parasitology , Genome , Oomycetes/classification , Oomycetes/pathogenicity , Phylogeny , Plants/parasitology , Saprolegnia/classification , Saprolegnia/pathogenicity
12.
Curr Genet ; 61(4): 567-77, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25754775

ABSTRACT

Phytophthora infestans is one of the most destructive plant pathogens of potato and tomato globally. The pathogen is closely related to four other Phytophthora species in the 1c clade including P. phaseoli, P. ipomoeae, P. mirabilis and P. andina that are important pathogens of other wild and domesticated hosts. P. andina is an interspecific hybrid between P. infestans and an unknown Phytophthora species. We have sequenced mitochondrial genomes of the sister species of P. infestans and examined the evolutionary relationships within the clade. Phylogenetic analysis indicates that the P. phaseoli mitochondrial lineage is basal within the clade. P. mirabilis and P. ipomoeae are sister lineages and share a common ancestor with the Ic mitochondrial lineage of P. andina. These lineages in turn are sister to the P. infestans and P. andina Ia mitochondrial lineages. The P. andina Ic lineage diverged much earlier than the P. andina Ia mitochondrial lineage and P. infestans. The presence of two mitochondrial lineages in P. andina supports the hybrid nature of this species. The ancestral state of the P. andina Ic lineage in the tree and its occurrence only in the Andean regions of Ecuador, Colombia and Peru suggests that the origin of this species hybrid in nature may occur there.


Subject(s)
Genome, Mitochondrial , Mitochondria/genetics , Phylogeny , Phytophthora infestans/genetics , Phytophthora/genetics , Biological Evolution , Chimera/microbiology , Colombia , DNA, Mitochondrial/genetics , Ecuador , Solanum lycopersicum/microbiology , Peru , Phylogeography , Phytophthora/classification , Phytophthora infestans/classification , Plant Diseases/microbiology , Sequence Analysis, DNA , Solanum tuberosum/microbiology
13.
Genome Res ; 22(11): 2270-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22829535

ABSTRACT

Exceptionally accurate genome reference sequences have proven to be of great value to microbial researchers. Thus, to date, about 1800 bacterial genome assemblies have been "finished" at great expense with the aid of manual laboratory and computational processes that typically iterate over a period of months or even years. By applying a new laboratory design and new assembly algorithm to 16 samples, we demonstrate that assemblies exceeding finished quality can be obtained from whole-genome shotgun data and automated computation. Cost and time requirements are thus dramatically reduced.


Subject(s)
Bacteria/genetics , Genome, Bacterial , Genomic Library , Sequence Analysis, DNA/methods , Algorithms
14.
Proc Natl Acad Sci U S A ; 109(8): 3065-70, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22315421

ABSTRACT

The degree to which molecular epidemiology reveals information about the sources and transmission patterns of an outbreak depends on the resolution of the technology used and the samples studied. Isolates of Escherichia coli O104:H4 from the outbreak centered in Germany in May-July 2011, and the much smaller outbreak in southwest France in June 2011, were indistinguishable by standard tests. We report a molecular epidemiological analysis using multiplatform whole-genome sequencing and analysis of multiple isolates from the German and French outbreaks. Isolates from the German outbreak showed remarkably little diversity, with only two single nucleotide polymorphisms (SNPs) found in isolates from four individuals. Surprisingly, we found much greater diversity (19 SNPs) in isolates from seven individuals infected in the French outbreak. The German isolates form a clade within the more diverse French outbreak strains. Moreover, five isolates derived from a single infected individual from the French outbreak had extremely limited diversity. The striking difference in diversity between the German and French outbreak samples is consistent with several hypotheses, including a bottleneck that purged diversity in the German isolates, variation in mutation rates in the two E. coli outbreak populations, or uneven distribution of diversity in the seed populations that led to each outbreak.


Subject(s)
Disease Outbreaks/statistics & numerical data , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/genetics , Europe/epidemiology , Humans , Models, Genetic , Phylogeny , Polymorphism, Single Nucleotide/genetics
15.
Nat Genet ; 38(4): 411-3, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16501576

ABSTRACT

We recently identified angiogenin (ANG) as a candidate susceptibility gene for amyotrophic lateral sclerosis (ALS), a neurodegenerative disorder characterized by adult-onset loss of motor neurons. We now report the finding of seven missense mutations in 15 individuals, of whom four had familial ALS and 11 apparently 'sporadic' ALS. Our findings provide further evidence that variations in hypoxia-inducible genes have an important role in motor neuron degeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Mutation, Missense , Ribonuclease, Pancreatic/genetics , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Models, Molecular , Pedigree , Protein Conformation , Ribonuclease, Pancreatic/chemistry
16.
J Biol Chem ; 287(1): 672-681, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22086920

ABSTRACT

Schizosaccharomyces pombe Sre1 is a membrane-bound transcription factor that controls adaptation to hypoxia. Like its mammalian homolog, sterol regulatory element-binding protein (SREBP), Sre1 activation requires release from the membrane. However, in fission yeast, this release occurs through a strikingly different mechanism that requires the Golgi Dsc E3 ubiquitin ligase complex and the proteasome. The mechanistic details of Sre1 cleavage, including the link between the Dsc E3 ligase complex and proteasome, are not well understood. Here, we present results of a genetic selection designed to identify additional components required for Sre1 cleavage. From the selection, we identified two new components of the fission yeast SREBP pathway: Dsc5 and Cdc48. The AAA (ATPase associated with diverse cellular activities) ATPase Cdc48 and Dsc5, a ubiquitin regulatory X domain-containing protein, interact with known Dsc complex components and are required for SREBP cleavage. These findings provide a mechanistic link between the Dsc E3 ligase complex and the proteasome in SREBP cleavage and add to a growing list of similarities between the Dsc E3 ligase and membrane E3 ligases involved in endoplasmic reticulum-associated degradation.


Subject(s)
Adenosine Triphosphatases/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Membrane Proteins/metabolism , Protein Subunits/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Carrier Proteins/chemistry , Endoplasmic Reticulum/metabolism , Membrane Proteins/chemistry , Mutagenesis , Protein Structure, Tertiary , Protein Subunits/chemistry , Proteolysis , Schizosaccharomyces/cytology , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/chemistry , Valosin Containing Protein
17.
Nature ; 448(7153): 553-60, 2007 Aug 02.
Article in English | MEDLINE | ID: mdl-17603471

ABSTRACT

We report the application of single-molecule-based sequencing technology for high-throughput profiling of histone modifications in mammalian cells. By obtaining over four billion bases of sequence from chromatin immunoprecipitated DNA, we generated genome-wide chromatin-state maps of mouse embryonic stem cells, neural progenitor cells and embryonic fibroblasts. We find that lysine 4 and lysine 27 trimethylation effectively discriminates genes that are expressed, poised for expression, or stably repressed, and therefore reflect cell state and lineage potential. Lysine 36 trimethylation marks primary coding and non-coding transcripts, facilitating gene annotation. Trimethylation of lysine 9 and lysine 20 is detected at satellite, telomeric and active long-terminal repeats, and can spread into proximal unique sequences. Lysine 4 and lysine 9 trimethylation marks imprinting control regions. Finally, we show that chromatin state can be read in an allele-specific manner by using single nucleotide polymorphisms. This study provides a framework for the application of comprehensive chromatin profiling towards characterization of diverse mammalian cell populations.


Subject(s)
Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , Genome/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Alleles , Animals , CpG Islands/genetics , Fibroblasts , Gene Expression Regulation, Developmental , Genomic Imprinting , Histones/metabolism , Male , Methylation , Mice , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics
18.
Nat Commun ; 14(1): 3907, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37400441

ABSTRACT

YAP is a key transcriptional co-activator of TEADs, it regulates cell growth and is frequently activated in cancer. In Malignant Pleural Mesothelioma (MPM), YAP is activated by loss-of-function mutations in upstream components of the Hippo pathway, while, in Uveal Melanoma (UM), YAP is activated in a Hippo-independent manner. To date, it is unclear if and how the different oncogenic lesions activating YAP impact its oncogenic program, which is particularly relevant for designing selective anti-cancer therapies. Here we show that, despite YAP being essential in both MPM and UM, its interaction with TEAD is unexpectedly dispensable in UM, limiting the applicability of TEAD inhibitors in this cancer type. Systematic functional interrogation of YAP regulatory elements in both cancer types reveals convergent regulation of broad oncogenic drivers in both MPM and UM, but also strikingly selective programs. Our work reveals unanticipated lineage-specific features of the YAP regulatory network that provide important insights to guide the design of tailored therapeutic strategies to inhibit YAP signaling across different cancer types.


Subject(s)
Adaptor Proteins, Signal Transducing , Neoplasms , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins , Epigenomics , Transcription Factors/genetics , Transcription Factors/metabolism , Signal Transduction/genetics
19.
BMC Genomics ; 13: 375, 2012 Aug 05.
Article in English | MEDLINE | ID: mdl-22863213

ABSTRACT

BACKGROUND: Pacific Biosciences technology provides a fundamentally new data type that provides the potential to overcome some limitations of current next generation sequencing platforms by providing significantly longer reads, single molecule sequencing, low composition bias and an error profile that is orthogonal to other platforms. With these potential advantages in mind, we here evaluate the utility of the Pacific Biosciences RS platform for human medical amplicon resequencing projects. RESULTS: We evaluated the Pacific Biosciences technology for SNP discovery in medical resequencing projects using the Genome Analysis Toolkit, observing high sensitivity and specificity for calling differences in amplicons containing known true or false SNPs. We assessed data quality: most errors were indels (~14%) with few apparent miscalls (~1%). In this work, we define a custom data processing pipeline for Pacific Biosciences data for human data analysis. CONCLUSION: Critically, the error properties were largely free of the context-specific effects that affect other sequencing technologies. These data show excellent utility for follow-up validation and extension studies in human data and medical genetics projects, but can be extended to other organisms with a reference genome.


Subject(s)
Sequence Analysis, DNA , Genetic Variation , Genome, Human , Genotype , Humans , Polymorphism, Single Nucleotide , Software , User-Computer Interface
20.
Nat Methods ; 6(1): 99-103, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19043412

ABSTRACT

Cancer results from somatic alterations in key genes, including point mutations, copy-number alterations and structural rearrangements. A powerful way to discover cancer-causing genes is to identify genomic regions that show recurrent copy-number alterations (gains and losses) in tumor genomes. Recent advances in sequencing technologies suggest that massively parallel sequencing may provide a feasible alternative to DNA microarrays for detecting copy-number alterations. Here we present: (i) a statistical analysis of the power to detect copy-number alterations of a given size; (ii) SegSeq, an algorithm to segment equal copy numbers from massively parallel sequence data; and (iii) analysis of experimental data from three matched pairs of tumor and normal cell lines. We show that a collection of approximately 14 million aligned sequence reads from human cell lines has comparable power to detect events as the current generation of DNA microarrays and has over twofold better precision for localizing breakpoints (typically, to within approximately 1 kilobase).


Subject(s)
Databases, Genetic , Gene Dosage/genetics , Algorithms , Base Sequence , Cell Line, Tumor , Chromosomes, Human/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL