Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Cell ; 83(14): 2478-2492.e8, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37369201

ABSTRACT

The RNA-binding protein TRIM71/LIN-41 is a phylogenetically conserved developmental regulator that functions in mammalian stem cell reprogramming, brain development, and cancer. TRIM71 recognizes target mRNAs through hairpin motifs and silences them through molecular mechanisms that await identification. Here, we uncover that TRIM71 represses its targets through RNA-supported interaction with TNRC6/GW182, a core component of the miRNA-induced silencing complex (miRISC). We demonstrate that AGO2, TRIM71, and UPF1 each recruit TNRC6 to specific sets of transcripts to silence them. As cellular TNRC6 levels are limiting, competition occurs among the silencing pathways, such that the loss of AGO proteins or of AGO binding to TNRC6 enhances the activities of the other pathways. We conclude that a miRNA-like silencing activity is shared among different mRNA silencing pathways and that the use of TNRC6 as a central hub provides a means to integrate their activities.


Subject(s)
Argonaute Proteins , MicroRNAs , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Protein Binding , Stem Cells/metabolism , Mammals/metabolism
2.
Nature ; 550(7674): 114-118, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28953874

ABSTRACT

The ability to directly uncover the contributions of genes to a given phenotype is fundamental for biology research. However, ostensibly homogeneous cell populations exhibit large clonal variance that can confound analyses and undermine reproducibility. Here we used genome-saturated mutagenesis to create a biobank of over 100,000 individual haploid mouse embryonic stem (mES) cell lines targeting 16,970 genes with genetically barcoded, conditional and reversible mutations. This Haplobank is, to our knowledge, the largest resource of hemi/homozygous mutant mES cells to date and is available to all researchers. Reversible mutagenesis overcomes clonal variance by permitting functional annotation of the genome directly in sister cells. We use the Haplobank in reverse genetic screens to investigate the temporal resolution of essential genes in mES cells, and to identify novel genes that control sprouting angiogenesis and lineage specification of blood vessels. Furthermore, a genome-wide forward screen with Haplobank identified PLA2G16 as a host factor that is required for cytotoxicity by rhinoviruses, which cause the common cold. Therefore, clones from the Haplobank combined with the use of reversible technologies enable high-throughput, reproducible, functional annotation of the genome.


Subject(s)
Biological Specimen Banks , Genomics/methods , Haploidy , Mouse Embryonic Stem Cells/metabolism , Mutation , Animals , Blood Vessels/cytology , Cell Lineage/genetics , Common Cold/genetics , Common Cold/virology , Genes, Essential/genetics , Genetic Testing , HEK293 Cells , Homozygote , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Neovascularization, Physiologic/genetics , Phospholipases A2, Calcium-Independent/genetics , Phospholipases A2, Calcium-Independent/metabolism , Rhinovirus/pathogenicity
3.
J Nat Prod ; 83(4): 965-971, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32182062

ABSTRACT

Kendomycin is a small-molecule natural product that has gained significant attention due to reported cytotoxicity against pathogenic bacteria and fungi as well as a number of cancer cell lines. Despite significant biomedical interest and attempts to reveal its mechanism of action, the cellular target of kendomycin remains disputed. Herein it is shown that kendomycin induces cellular responses indicative of cation stress comparable to the effects of established iron chelators. Furthermore, addition of excess iron and copper attenuated kendomycin cytotoxicity in bacteria, yeast, and mammalian cells. Finally, NMR analysis demonstrated a direct interaction with cations, corroborating a close link between the observed kendomycin polypharmacology across different species and modulation of iron and/or copper levels.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antifungal Agents/pharmacology , Bacteria/drug effects , Chelating Agents/pharmacology , Fungi/drug effects , Rifabutin/analogs & derivatives , Cations , Cell Line , Copper/metabolism , Iron/metabolism , Leupeptins/pharmacology , Microbial Sensitivity Tests , Mutagenesis , Rifabutin/pharmacology , Yeasts/drug effects
4.
J Cell Sci ; 128(6): 1217-29, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25616894

ABSTRACT

A new cyclic decadepsipeptide was isolated from Chaetosphaeria tulasneorum with potent bioactivity on mammalian and yeast cells. Chemogenomic profiling in S. cerevisiae indicated that the Sec61 translocon complex, the machinery for protein translocation and membrane insertion at the endoplasmic reticulum, is the target. The profiles were similar to those of cyclic heptadepsipeptides of a distinct chemotype (including HUN-7293 and cotransin) that had previously been shown to inhibit cotranslational translocation at the mammalian Sec61 translocon. Unbiased, genome-wide mutagenesis followed by full-genome sequencing in both fungal and mammalian cells identified dominant mutations in Sec61p (yeast) or Sec61α1 (mammals) that conferred resistance. Most, but not all, of these mutations affected inhibition by both chemotypes, despite an absence of structural similarity. Biochemical analysis confirmed inhibition of protein translocation into the endoplasmic reticulum of both co- and post-translationally translocated substrates by both chemotypes, demonstrating a mechanism independent of a translating ribosome. Most interestingly, both chemotypes were found to also inhibit SecYEG, the bacterial Sec61 translocon homolog. We suggest 'decatransin' as the name for this new decadepsipeptide translocation inhibitor.


Subject(s)
Biological Products/pharmacology , Endoplasmic Reticulum/drug effects , Membrane Proteins/metabolism , Protein Transport/drug effects , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Ascomycota/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , HCT116 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Polymorphism, Single Nucleotide/genetics , SEC Translocation Channels , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development
5.
Angew Chem Int Ed Engl ; 54(35): 10149-54, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26179970

ABSTRACT

Cultivation of myxobacteria of the Nannocystis genus led to the isolation and structure elucidation of a class of novel cyclic lactone inhibitors of elongation factor 1. Whole genome sequence analysis and annotation enabled identification of the putative biosynthetic cluster and synthesis process. In biological assays the compounds displayed anti-fungal and cytotoxic activity. Combined genetic and proteomic approaches identified the eukaryotic translation elongation factor 1α (EF-1α) as the primary target for this compound class. Nannocystin A (1) displayed differential activity across various cancer cell lines and EEF1A1 expression levels appear to be the main differentiating factor. Biochemical and genetic evidence support an overlapping binding site of 1 with the anti-cancer compound didemnin B on EF-1α. This myxobacterial chemotype thus offers an interesting starting point for further investigations of the potential of therapeutics targeting elongation factor 1.


Subject(s)
Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Macrocyclic Compounds/pharmacology , Myxococcales/physiology , Neoplasms/pathology , Peptide Elongation Factor 1/antagonists & inhibitors , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Candida albicans/drug effects , Genomics/methods , Humans , Macrocyclic Compounds/chemistry , Molecular Structure , Neoplasms/drug therapy , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Proteomics/methods , Structure-Activity Relationship , Tumor Cells, Cultured
6.
bioRxiv ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38712281

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) - characterized by excess accumulation of fat in the liver - now affects one third of the world's population. As NAFLD progresses, extracellular matrix components including collagen accumulate in the liver causing tissue fibrosis, a major determinant of disease severity and mortality. To identify transcriptional regulators of fibrosis, we computationally inferred the activity of transcription factors (TFs) relevant to fibrosis by profiling the matched transcriptomes and epigenomes of 108 human liver biopsies from a deeply-characterized cohort of patients spanning the full histopathologic spectrum of NAFLD. CRISPR-based genetic knockout of the top 100 TFs identified ZNF469 as a regulator of collagen expression in primary human hepatic stellate cells (HSCs). Gain- and loss-of-function studies established that ZNF469 regulates collagen genes and genes involved in matrix homeostasis through direct binding to gene bodies and regulatory elements. By integrating multiomic large-scale profiling of human biopsies with extensive experimental validation we demonstrate that ZNF469 is a transcriptional regulator of collagen in HSCs. Overall, these data nominate ZNF469 as a previously unrecognized determinant of NAFLD-associated liver fibrosis.

7.
Antimicrob Agents Chemother ; 57(5): 2272-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23478965

ABSTRACT

High-throughput phenotypic screening against the yeast Saccharomyces cerevisiae revealed a series of triazolopyrimidine-sulfonamide compounds with broad-spectrum antifungal activity, no significant cytotoxicity, and low protein binding. To elucidate the target of this series, we have applied a chemogenomic profiling approach using the S. cerevisiae deletion collection. All compounds of the series yielded highly similar profiles that suggested acetolactate synthase (Ilv2p, which catalyzes the first common step in branched-chain amino acid biosynthesis) as a possible target. The high correlation with profiles of known Ilv2p inhibitors like chlorimuron-ethyl provided further evidence for a similar mechanism of action. Genome-wide mutagenesis in S. cerevisiae identified 13 resistant clones with 3 different mutations in the catalytic subunit of acetolactate synthase that also conferred cross-resistance to established Ilv2p inhibitors. Mapping of the mutations into the published Ilv2p crystal structure outlined the chlorimuron-ethyl binding cavity, and it was possible to dock the triazolopyrimidine-sulfonamide compound into this pocket in silico. However, fungal growth inhibition could be bypassed through supplementation with exogenous branched-chain amino acids or by the addition of serum to the medium in all of the fungal organisms tested except for Aspergillus fumigatus. Thus, these data support the identification of the triazolopyrimidine-sulfonamide compounds as inhibitors of acetolactate synthase but suggest that targeting may be compromised due to the possibility of nutrient bypass in vivo.


Subject(s)
Acetolactate Synthase/antagonists & inhibitors , Antifungal Agents/pharmacology , Pyrimidines/pharmacology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Sulfonamides/pharmacology , Sulfonylurea Compounds/pharmacology , Acetolactate Synthase/chemistry , Acetolactate Synthase/genetics , Acetolactate Synthase/metabolism , Amino Acids, Branched-Chain/metabolism , Amino Acids, Branched-Chain/pharmacology , Antifungal Agents/chemistry , Catalytic Domain/drug effects , High-Throughput Screening Assays , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutation , Protein Binding , Pyrimidines/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Serum/chemistry , Serum/metabolism , Sulfonamides/chemistry , Sulfonylurea Compounds/chemistry
8.
iScience ; 24(11): 103323, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34805786

ABSTRACT

BET bromodomain inhibitors hold promise as therapeutic agents in diverse indications, but their clinical progression has been challenging and none have received regulatory approval. Early clinical trials in cancer have shown heterogeneous clinical responses, development of resistance, and adverse events. Increased understanding of their mechanism(s) of action and identification of biomarkers are needed to identify appropriate indication(s) and achieve efficacious dosing. Using genome-wide CRISPR-Cas9 screens at different concentrations, we report molecular mechanisms defining cellular responses to BET inhibitors, some of which appear specific to a single compound concentration. We identify multiple transcriptional regulators and mTOR pathway members as key determinants of JQ1 sensitivity and two Ca2+/Mn2+ transporters, ATP2C1 and TMEM165, as key determinants of JQ1 resistance. Our study reveals new molecular mediators of BET bromodomain inhibitor effects, suggests the involvement of manganese, and provides a rich resource for discovery of biomarkers and targets for combination therapies.

10.
Nat Commun ; 11(1): 3387, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636417

ABSTRACT

Biosynthesis of glycosylphosphatidylinositol (GPI) is required for anchoring proteins to the plasma membrane, and is essential for the integrity of the fungal cell wall. Here, we use a reporter gene-based screen in Saccharomyces cerevisiae for the discovery of antifungal inhibitors of GPI-anchoring of proteins, and identify the oligocyclopropyl-containing natural product jawsamycin (FR-900848) as a potent hit. The compound targets the catalytic subunit Spt14 (also referred to as Gpi3) of the fungal UDP-glycosyltransferase, the first step in GPI biosynthesis, with good selectivity over the human functional homolog PIG-A. Jawsamycin displays antifungal activity in vitro against several pathogenic fungi including Mucorales, and in vivo in a mouse model of invasive pulmonary mucormycosis due to Rhyzopus delemar infection. Our results provide a starting point for the development of Spt14 inhibitors for treatment of invasive fungal infections.


Subject(s)
Antifungal Agents/pharmacology , Glycosyltransferases/antagonists & inhibitors , Polyketides/pharmacology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Animals , Cell Proliferation , Disease Models, Animal , Fermentation , Genes, Reporter , Glycosylphosphatidylinositols/biosynthesis , HCT116 Cells , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , K562 Cells , Lung/microbiology , Male , Mice , Mice, Inbred ICR , Mucorales , Multigene Family , Rhizopus , Saccharomyces cerevisiae
11.
Nat Commun ; 10(1): 4676, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31611556

ABSTRACT

Resident adult epithelial stem cells maintain tissue homeostasis by balancing self-renewal and differentiation. The stem cell potential of human epidermal keratinocytes is retained in vitro but lost over time suggesting extrinsic and intrinsic regulation. Transcription factor-controlled regulatory circuitries govern cell identity, are sufficient to induce pluripotency and transdifferentiate cells. We investigate whether transcriptional circuitry also governs phenotypic changes within a given cell type by comparing human primary keratinocytes with intrinsically high versus low stem cell potential. Using integrated chromatin and transcriptional profiling, we implicate IRF2 as antagonistic to stemness and show that it binds and regulates active cis-regulatory elements at interferon response and antigen presentation genes. CRISPR-KD of IRF2 in keratinocytes with low stem cell potential increases self-renewal, migration and epidermis formation. These data demonstrate that transcription factor regulatory circuitries, in addition to maintaining cell identity, control plasticity within cell types and offer potential for therapeutic modulation of cell function.


Subject(s)
Interferon Regulatory Factor-2/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Humans , Interferon Regulatory Factor-2/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Transcriptional Activation/physiology
12.
ACS Chem Biol ; 12(9): 2264-2269, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28796488

ABSTRACT

The microbial metabolite Chivosazole F has been described to affect the cytoskeleton and to inhibit actin polymerization in vitro. Applying orthogonal genomic and proteomics approaches, we now show for the first time that Chivosazole F exerts its effect by directly interacting with actin and demonstrate the cellular impact of Chivosazole F in an unbiased, genome-wide context in yeast and in mammalian cells. Furthermore, mutation-based resistance mapping identifies two SNPs located in the putative Chivosazole F binding site of actin. Comparing chemogenomic profiles and responses to the Chivosazole F-resistant SNPs shows a partially conserved mechanism of action for Chivosazole F and Latrunculin A, but clear divergence from Chondramide. In addition, C14orf80 is an evolutionarily highly conserved ORF, lacking any functional annotation. As editing of C14orf80 leads to Chivosazole F hyper-resistance, we propose a function for this gene product in counteracting perturbation of actin filaments.


Subject(s)
Actins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Depsipeptides/pharmacology , Macrolides/pharmacology , Thiazolidines/pharmacology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actins/genetics , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Depsipeptides/chemistry , HEK293 Cells , Humans , Macrolides/chemistry , Mutation , Myxococcales/chemistry , Thiazolidines/chemistry
13.
Cell Rep ; 19(3): 451-460, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423309

ABSTRACT

Flavivirus infections by Zika and dengue virus impose a significant global healthcare threat with no US Food and Drug Administration (FDA)-approved vaccination or specific antiviral treatment available. Here, we present the discovery of an anti-flaviviral natural product named cavinafungin. Cavinafungin is a potent and selectively active compound against Zika and all four dengue virus serotypes. Unbiased, genome-wide genomic profiling in human cells using a novel CRISPR/Cas9 protocol identified the endoplasmic-reticulum-localized signal peptidase as the efficacy target of cavinafungin. Orthogonal profiling in S. cerevisiae followed by the selection of resistant mutants pinpointed the catalytic subunit of the signal peptidase SEC11 as the evolutionary conserved target. Biochemical analysis confirmed a rapid block of signal sequence cleavage of both host and viral proteins by cavinafungin. This study provides an effective compound against the eukaryotic signal peptidase and independent confirmation of the recently identified critical role of the signal peptidase in the replicative cycle of flaviviruses.


Subject(s)
Biological Products/pharmacology , Dengue Virus/physiology , Lipopeptides/pharmacology , Virus Replication/drug effects , Zika Virus/physiology , Biological Products/chemistry , CRISPR-Cas Systems/genetics , Dengue Virus/drug effects , Gene Knockdown Techniques , Genome, Human , Genomics , HCT116 Cells , Humans , Lipopeptides/chemistry , Membrane Proteins , Protein Subunits/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Serine Endopeptidases , Viral Proteins/metabolism , Zika Virus/drug effects
14.
Sci Rep ; 7: 42728, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28205648

ABSTRACT

Chemogenomic profiling is a powerful and unbiased approach to elucidate pharmacological targets and the mechanism of bioactive compounds. Until recently, genome-wide, high-resolution experiments of this nature have been limited to fungal systems due to lack of mammalian genome-wide deletion collections. With the example of a novel nicotinamide phosphoribosyltransferase (NAMPT) inhibitor, we demonstrate that the CRISPR/Cas9 system enables the generation of transient homo- and heterozygous deletion libraries and allows for the identification of efficacy targets and pathways mediating hypersensitivity and resistance relevant to the compound mechanism of action.


Subject(s)
CRISPR-Cas Systems , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Cells, Cultured , Enzyme Inhibitors/chemistry , Gene Deletion , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Pharmacogenomic Testing/methods
15.
Microbiol Res ; 169(2-3): 107-20, 2014.
Article in English | MEDLINE | ID: mdl-24360837

ABSTRACT

Due to evolutionary conservation of biology, experimental knowledge captured from genetic studies in eukaryotic model organisms provides insight into human cellular pathways and ultimately physiology. Yeast chemogenomic profiling is a powerful approach for annotating cellular responses to small molecules. Using an optimized platform, we provide the relative sensitivities of the heterozygous and homozygous deletion collections for nearly 1800 biologically active compounds. The data quality enables unique insights into pathways that are sensitive and resistant to a given perturbation, as demonstrated with both known and novel compounds. We present examples of novel compounds that inhibit the therapeutically relevant fatty acid synthase and desaturase (Fas1p and Ole1p), and demonstrate how the individual profiles facilitate hypothesis-driven experiments to delineate compound mechanism of action. Importantly, the scale and diversity of tested compounds yields a dataset where the number of modulated pathways approaches saturation. This resource can be used to map novel biological connections, and also identify functions for unannotated genes. We validated hypotheses generated by global two-way hierarchical clustering of profiles for (i) novel compounds with a similar mechanism of action acting upon microtubules or vacuolar ATPases, and (ii) an un-annotated ORF, YIL060w, that plays a role in respiration in the mitochondria. Finally, we identify and characterize background mutations in the widely used yeast deletion collection which should improve the interpretation of past and future screens throughout the community. This comprehensive resource of cellular responses enables the expansion of our understanding of eukaryotic pathway biology.


Subject(s)
Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Antifungal Agents/pharmacology , Biosynthetic Pathways , Drug Resistance, Fungal , Gene Expression Regulation, Fungal , High-Throughput Screening Assays , Molecular Sequence Data , Phylogeny , Saccharomyces cerevisiae/classification , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/metabolism
16.
PLoS One ; 7(9): e42657, 2012.
Article in English | MEDLINE | ID: mdl-22970117

ABSTRACT

Argyrins, produced by myxobacteria and actinomycetes, are cyclic octapeptides with antibacterial and antitumor activity. Here, we identify elongation factor G (EF-G) as the cellular target of argyrin B in bacteria, via resistant mutant selection and whole genome sequencing, biophysical binding studies and crystallography. Argyrin B binds a novel allosteric pocket in EF-G, distinct from the known EF-G inhibitor antibiotic fusidic acid, revealing a new mode of protein synthesis inhibition. In eukaryotic cells, argyrin B was found to target mitochondrial elongation factor G1 (EF-G1), the closest homologue of bacterial EF-G. By blocking mitochondrial translation, argyrin B depletes electron transport components and inhibits the growth of yeast and tumor cells. Further supporting direct inhibition of EF-G1, expression of an argyrin B-binding deficient EF-G1 L693Q variant partially rescued argyrin B-sensitivity in tumor cells. In summary, we show that argyrin B is an antibacterial and cytotoxic agent that inhibits the evolutionarily conserved target EF-G, blocking protein synthesis in bacteria and mitochondrial translation in yeast and mammalian cells.


Subject(s)
Oligopeptides/metabolism , Peptide Elongation Factor G/metabolism , Allosteric Site , Amino Acid Sequence , Animals , Burkholderia/drug effects , Cell Line, Tumor , Conserved Sequence , Crystallography, X-Ray , Humans , Mammals , Microbial Sensitivity Tests , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptide Elongation Factor G/antagonists & inhibitors , Peptide Elongation Factor G/chemistry , Protein Binding/drug effects , Pseudomonas aeruginosa/drug effects , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL