Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cell ; 184(1): 76-91.e13, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33147444

ABSTRACT

Identification of host genes essential for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may reveal novel therapeutic targets and inform our understanding of coronavirus disease 2019 (COVID-19) pathogenesis. Here we performed genome-wide CRISPR screens in Vero-E6 cells with SARS-CoV-2, Middle East respiratory syndrome CoV (MERS-CoV), bat CoV HKU5 expressing the SARS-CoV-1 spike, and vesicular stomatitis virus (VSV) expressing the SARS-CoV-2 spike. We identified known SARS-CoV-2 host factors, including the receptor ACE2 and protease Cathepsin L. We additionally discovered pro-viral genes and pathways, including HMGB1 and the SWI/SNF chromatin remodeling complex, that are SARS lineage and pan-coronavirus specific, respectively. We show that HMGB1 regulates ACE2 expression and is critical for entry of SARS-CoV-2, SARS-CoV-1, and NL63. We also show that small-molecule antagonists of identified gene products inhibited SARS-CoV-2 infection in monkey and human cells, demonstrating the conserved role of these genetic hits across species. This identifies potential therapeutic targets for SARS-CoV-2 and reveals SARS lineage-specific and pan-CoV host factors that regulate susceptibility to highly pathogenic CoVs.


Subject(s)
Coronavirus Infections/genetics , Genome-Wide Association Study , Host-Pathogen Interactions , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/immunology , COVID-19/virology , Cell Line , Chlorocebus aethiops , Clustered Regularly Interspaced Short Palindromic Repeats , Coronavirus/classification , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Gene Knockout Techniques , Gene Regulatory Networks , HEK293 Cells , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Host-Pathogen Interactions/drug effects , Humans , Vero Cells , Virus Internalization
2.
PLoS Biol ; 22(7): e3002718, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38976757

ABSTRACT

Cancer initiates as a consequence of genomic mutations and its subsequent progression relies in part on increased production of ribosomes to maintain high levels of protein synthesis for unchecked cell growth. Recently, cytidine deaminases have been uncovered as sources of mutagenesis in cancer. In an attempt to form a connection between these 2 cancer driving processes, we interrogated the cytidine deaminase family of proteins for potential roles in human ribosome biogenesis. We identified and validated APOBEC3A and APOBEC4 as novel ribosome biogenesis factors through our laboratory's established screening platform for the discovery of regulators of nucleolar function in MCF10A cells. Through siRNA depletion experiments, we highlight APOBEC3A's requirement in making ribosomes and specific role within the processing and maturation steps that form the large subunit 5.8S and 28S ribosomal (r)RNAs. We demonstrate that a subset of APOBEC3A resides within the nucleolus and associates with critical ribosome biogenesis factors. Mechanistic insight was revealed by transient overexpression of both wild-type and a catalytically dead mutated APOBEC3A, which both increase cell growth and protein synthesis. Through an innovative nuclear RNA sequencing methodology, we identify only modest predicted APOBEC3A C-to-U target sites on the pre-rRNA and pre-mRNAs. Our work reveals a potential direct role for APOBEC3A in ribosome biogenesis likely independent of its editing function. More broadly, we found an additional function of APOBEC3A in cancer pathology through its function in ribosome biogenesis, expanding its relevance as a target for cancer therapeutics.


Subject(s)
Cell Nucleolus , Cell Proliferation , Cytidine Deaminase , Ribosomes , Humans , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Cell Nucleolus/metabolism , Ribosomes/metabolism , Cell Proliferation/genetics , RNA, Ribosomal/metabolism , RNA, Ribosomal/genetics , Cell Line, Tumor , Proteins/metabolism , Proteins/genetics
3.
Nucleic Acids Res ; 52(4): 1988-2011, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38197221

ABSTRACT

While microRNAs and other non-coding RNAs are the next frontier of novel regulators of mammalian ribosome biogenesis (RB), a systematic exploration of microRNA-mediated RB regulation has not yet been undertaken. We carried out a high-content screen in MCF10A cells for changes in nucleolar number using a library of 2603 mature human microRNA mimics. Following a secondary screen for nucleolar rRNA biogenesis inhibition, we identified 72 novel microRNA negative regulators of RB after stringent hit calling. Hits included 27 well-conserved microRNAs present in MirGeneDB, and were enriched for mRNA targets encoding proteins with nucleolar localization or functions in cell cycle regulation. Rigorous selection and validation of a subset of 15 microRNA hits unexpectedly revealed that most of them caused dysregulated pre-rRNA processing, elucidating a novel role for microRNAs in RB regulation. Almost all hits impaired global protein synthesis and upregulated CDKN1A (p21) levels, while causing diverse effects on RNA Polymerase 1 (RNAP1) transcription and TP53 protein levels. We provide evidence that the MIR-28 siblings, hsa-miR-28-5p and hsa-miR-708-5p, potently target the ribosomal protein mRNA RPS28 via tandem primate-specific 3' UTR binding sites, causing a severe pre-18S pre-rRNA processing defect. Our work illuminates novel microRNA attenuators of RB, forging a promising new path for microRNA mimic chemotherapeutics.


Subject(s)
MicroRNAs , RNA Precursors , Ribosomes , Animals , Humans , Mammals/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Ribosomes/genetics , Ribosomes/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/metabolism , Transcription Factors/metabolism
4.
RNA ; 2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36323459

ABSTRACT

In eukaryotes, the nucleolus is the site of ribosome biosynthesis, an essential process in all cells. While human ribosome assembly is largely evolutionarily conserved, many of the regulatory details underlying its control and function have not yet been well-defined. The nucleolar protein RSL24D1 was originally identified as a factor important for 60S ribosomal subunit biogenesis. In addition, the PeBoW (BOP1-PES1-WDR12) complex has been well-defined as required for pre-28S rRNA processing and cell proliferation. In this study, we show that RSL24D1 depletion impairs both pre-ribosomal RNA (pre-rRNA) transcription and mature 28S rRNA production, leading to decreased protein synthesis and p53 stabilization in human cells. Surprisingly, each of the PeBoW complex members is also required for pre-rRNA transcription. We demonstrate that RSL24D1 and WDR12 co-immunoprecipitate with the RNA polymerase I subunit, RPA194, and regulate its steady state levels. These results uncover the dual role of RSL24D1 and the PeBoW complex in multiple steps of ribosome biogenesis, and provide evidence implicating large ribosomal subunit biogenesis factors in pre-rRNA transcription control.

5.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Article in English | MEDLINE | ID: mdl-34185680

ABSTRACT

Translation of open reading frame 1b (ORF1b) in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires a programmed -1 ribosomal frameshift (-1 PRF) promoted by an RNA pseudoknot. The extent to which SARS-CoV-2 replication may be sensitive to changes in -1 PRF efficiency is currently unknown. Through an unbiased, reporter-based high-throughput compound screen, we identified merafloxacin, a fluoroquinolone antibacterial, as a -1 PRF inhibitor for SARS-CoV-2. Frameshift inhibition by merafloxacin is robust to mutations within the pseudoknot region and is similarly effective on -1 PRF of other betacoronaviruses. Consistent with the essential role of -1 PRF in viral gene expression, merafloxacin impedes SARS-CoV-2 replication in Vero E6 cells, thereby providing proof-of-principle for targeting -1 PRF as a plausible and effective antiviral strategy for SARS-CoV-2 and other coronaviruses.


Subject(s)
Antiviral Agents/pharmacology , Frameshifting, Ribosomal/drug effects , SARS-CoV-2/drug effects , Virus Replication/drug effects , Animals , Betacoronavirus , Chlorocebus aethiops , Fluoroquinolones/pharmacology , Frameshifting, Ribosomal/genetics , Mutation , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/genetics , SARS-CoV-2/physiology , Vero Cells
6.
RNA Biol ; 20(1): 257-271, 2023 01.
Article in English | MEDLINE | ID: mdl-37246770

ABSTRACT

The main components of the essential cellular process of eukaryotic ribosome biogenesis are highly conserved from yeast to humans. Among these, the U3 Associated Proteins (UTPs) are a small subunit processome subcomplex that coordinate the first two steps of ribosome biogenesis in transcription and pre-18S processing. While we have identified the human counterparts of most of the yeast Utps, the homologs of yeast Utp9 and Bud21 (Utp16) have remained elusive. In this study, we find that NOL7 is the likely ortholog of Bud21. Previously described as a tumour suppressor through regulation of antiangiogenic transcripts, we now show that NOL7 is required for early pre-rRNA accumulation and pre-18S rRNA processing in human cells. These roles lead to decreased protein synthesis and induction of the nucleolar stress response upon NOL7 depletion. Beyond Bud21's nonessential role in yeast, we establish human NOL7 as an essential UTP that is necessary to maintain both early pre-rRNA levels and processing.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 18S/metabolism , RNA, Small Nucleolar/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
7.
J Am Chem Soc ; 143(38): 15824-15833, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34524796

ABSTRACT

Colibactin is a genotoxic metabolite produced by commensal-pathogenic members of the human microbiome that possess the clb (aka pks) biosynthetic gene cluster. clb+ bacteria induce tumorigenesis in models of intestinal inflammation and have been causally linked to oncogenesis in humans. While colibactin is believed underlie these effects, it has not been possible to study the molecule directly due to its instability. Herein, we report the synthesis and biological studies of colibactin 742 (4), a stable colibactin derivative. We show that colibactin 742 (4) induces DNA interstrand-cross-links, activation of the Fanconi Anemia DNA repair pathway, and G2/M arrest in a manner similar to clb+E. coli. The linear precursor 9, which mimics the biosynthetic precursor to colibactin, also recapitulates the bacterial phenotype. In the course of this work, we discovered a novel cyclization pathway that was previously undetected in MS-based studies of colibactin, suggesting a refinement to the natural product structure and its mode of DNA binding. Colibactin 742 (4) and its precursor 9 will allow researchers to study colibactin's genotoxic effects independent of the producing organism for the first time.


Subject(s)
Escherichia coli Proteins/chemical synthesis , Peptides/chemical synthesis , Polyketides/chemical synthesis , DNA/chemistry , Escherichia coli/genetics , Humans , Microbiota/genetics , Molecular Conformation , Multigene Family , Mutagens/metabolism , Mutation , Oxidation-Reduction , Phenotype , Protein Binding , Structure-Activity Relationship
8.
Proc Natl Acad Sci U S A ; 115(38): E8996-E9005, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30181290

ABSTRACT

Insulin resistance drives the development of type 2 diabetes (T2D). In liver, diacylglycerol (DAG) is a key mediator of lipid-induced insulin resistance. DAG activates protein kinase C ε (PKCε), which phosphorylates and inhibits the insulin receptor. In rats, a 3-day high-fat diet produces hepatic insulin resistance through this mechanism, and knockdown of hepatic PKCε protects against high-fat diet-induced hepatic insulin resistance. Here, we employed a systems-level approach to uncover additional signaling pathways involved in high-fat diet-induced hepatic insulin resistance. We used quantitative phosphoproteomics to map global in vivo changes in hepatic protein phosphorylation in chow-fed, high-fat-fed, and high-fat-fed with PKCε knockdown rats to distinguish the impact of lipid- and PKCε-induced protein phosphorylation. This was followed by a functional siRNA-based screen to determine which dynamically regulated phosphoproteins may be involved in canonical insulin signaling. Direct PKCε substrates were identified by motif analysis of phosphoproteomics data and validated using a large-scale in vitro kinase assay. These substrates included the p70S6K substrates RPS6 and IRS1, which suggested cross talk between PKCε and p70S6K in high-fat diet-induced hepatic insulin resistance. These results identify an expanded set of proteins through which PKCε may drive high-fat diet-induced hepatic insulin resistance that may direct new therapeutic approaches for T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Protein Kinase C-epsilon/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Animals, Genetically Modified , Diabetes Mellitus, Type 2/etiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Knockdown Techniques , Humans , Insulin Receptor Substrate Proteins/metabolism , Lipid Metabolism/physiology , Liver/metabolism , Phosphorylation , Protein Kinase C-epsilon/genetics , Proteomics/methods , RNA, Small Interfering/metabolism , Rats , Receptor, Insulin/metabolism , Ribosomal Protein S6/metabolism , Signal Transduction/physiology
9.
J Biol Chem ; 294(32): 12146-12156, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31227523

ABSTRACT

Phosphatidylserine decarboxylases (PSDs) catalyze the decarboxylation of phosphatidylserine to generate phosphatidylethanolamine, a critical step in phospholipid metabolism in both prokaryotes and eukaryotes. Most PSDs are membrane-bound, and classical radioisotope-based assays for determining their activity in vitro are not suitable for high-throughput drug screening. The finding that the PkPSD from Plasmodium knowlesi can be purified in a soluble and active form and the recent development of a fluorescence-based distyrylbenzene-bis-aldehyde (DSB-3) assay to measure PSD activity in vitro have laid the groundwork for screening chemical libraries for PSD inhibitors. Using this assay, here we conducted a high-throughput screen of a structurally diverse 130,858-compound library against PkPSD. Further characterization of the hits identified in this screening yielded five PkPSD inhibitors with IC50 values ranging from 3.1 to 42.3 µm Lead compounds were evaluated against the pathogenic yeast Candida albicans in the absence or presence of exogenous ethanolamine, and YU253467 and YU254403 were identified as inhibiting both native C. albicans PSD mitochondrial activity and C. albicans growth, with an MIC50 of 22.5 and 15 µg/ml without ethanolamine and an MIC50 of 75 and 60 µg/ml with ethanolamine, respectively. Together, these results provide the first proof of principle for the application of DSB-3-based fluorescent readouts in high-throughput screening for PSD inhibitors. The data set the stage for future analyses to identify more selective and potent PSD inhibitors with antimicrobial or antitumor activities.


Subject(s)
Carboxy-Lyases/antagonists & inhibitors , Enzyme Inhibitors/analysis , Fluorescent Dyes/chemistry , High-Throughput Screening Assays , Styrenes/chemistry , Candida albicans/drug effects , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Ethanolamine/pharmacology , Humans , Inhibitory Concentration 50 , Phosphatidylserines/metabolism , Plasmodium knowlesi/enzymology , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification
10.
Proc Natl Acad Sci U S A ; 114(21): 5503-5508, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28500274

ABSTRACT

Cerebral cavernous malformations (CCMs) are common vascular anomalies that develop in the central nervous system and, more rarely, the retina. The lesions can cause headache, seizures, focal neurological deficits, and hemorrhagic stroke. Symptomatic lesions are treated according to their presentation; however, targeted pharmacological therapies that improve the outcome of CCM disease are currently lacking. We performed a high-throughput screen to identify Food and Drug Administration-approved drugs or other bioactive compounds that could effectively suppress hyperproliferation of mouse brain primary astrocytes deficient for CCM3. We demonstrate that fluvastatin, an inhibitor of 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase and the N-bisphosphonate zoledronic acid monohydrate, an inhibitor of protein prenylation, act synergistically to reverse outcomes of CCM3 loss in cultured mouse primary astrocytes and in Drosophila glial cells in vivo. Further, the two drugs effectively attenuate neural and vascular deficits in chronic and acute mouse models of CCM3 loss in vivo, significantly reducing lesion burden and extending longevity. Sustained inhibition of the mevalonate pathway represents a potential pharmacological treatment option and suggests advantages of combination therapy for CCM disease.


Subject(s)
Diphosphonates/therapeutic use , Fatty Acids, Monounsaturated/therapeutic use , Hemangioma, Cavernous, Central Nervous System/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Imidazoles/therapeutic use , Indoles/therapeutic use , Animals , Astrocytes/drug effects , Diphosphonates/pharmacology , Drosophila , Drug Evaluation, Preclinical , Drug Therapy, Combination , Endothelial Cells/drug effects , Female , Fluvastatin , High-Throughput Screening Assays , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mice , Pregnancy , Protein Prenylation/drug effects , Zoledronic Acid
11.
J Biol Chem ; 293(5): 1493-1503, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29247006

ABSTRACT

Phosphatidylserine decarboxylases (PSDs) are central enzymes in phospholipid metabolism that produce phosphatidylethanolamine (PE) in bacteria, protists, plants, and animals. We developed a fluorescence-based assay for selectively monitoring production of PE in reactions using a maltose-binding protein fusion with Plasmodium knowlesi PSD (MBP-His6-Δ34PkPSD) as the enzyme. The PE detection by fluorescence (λex = 403 nm, λem = 508 nm) occurred after the lipid reacted with a water-soluble distyrylbenzene-bis-aldehyde (DSB-3), and provided strong discrimination against the phosphatidylserine substrate. The reaction conditions were optimized for enzyme, substrate, product, and DSB-3 concentrations with the purified enzyme and also tested with crude extracts and membrane fractions from bacteria and yeast. The assay is readily amenable to application in 96- and 384-well microtiter plates and should prove useful for high-throughput screening for inhibitors of PSD enzymes across diverse phyla.


Subject(s)
Carboxy-Lyases/chemistry , Fluorescence , Maltose-Binding Proteins/chemistry , Phosphatidylethanolamines/chemistry , Plasmodium knowlesi/enzymology , Protozoan Proteins/chemistry , Carboxy-Lyases/genetics , Maltose-Binding Proteins/genetics , Phosphatidylethanolamines/analysis , Plasmodium knowlesi/genetics , Protozoan Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
12.
Proc Natl Acad Sci U S A ; 112(31): 9686-91, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195760

ABSTRACT

Complement membrane attack complexes (MACs) promote inflammatory functions in endothelial cells (ECs) by stabilizing NF-κB-inducing kinase (NIK) and activating noncanonical NF-κB signaling. Here we report a novel endosome-based signaling complex induced by MACs to stabilize NIK. We found that, in contrast to cytokine-mediated activation, NIK stabilization by MACs did not involve cIAP2 or TRAF3. Informed by a genome-wide siRNA screen, instead this response required internalization of MACs in a clathrin-, AP2-, and dynamin-dependent manner into Rab5(+)endosomes, which recruited activated Akt, stabilized NIK, and led to phosphorylation of IκB kinase (IKK)-α. Active Rab5 was required for recruitment of activated Akt to MAC(+) endosomes, but not for MAC internalization or for Akt activation. Consistent with these in vitro observations, MAC internalization occurred in human coronary ECs in vivo and was similarly required for NIK stabilization and EC activation. We conclude that MACs activate noncanonical NF-κB by forming a novel Akt(+)NIK(+) signalosome on Rab5(+) endosomes.


Subject(s)
Complement Membrane Attack Complex/metabolism , Endosomes/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , rab5 GTP-Binding Proteins/metabolism , Animals , Baculoviral IAP Repeat-Containing 3 Protein , Clathrin/metabolism , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endocytosis/drug effects , Endosomes/drug effects , Enzyme Stability/drug effects , Flow Cytometry , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrazones/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Mice, SCID , Protein Biosynthesis/drug effects , RNA, Small Interfering/metabolism , Secretory Vesicles/drug effects , Secretory Vesicles/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/metabolism , NF-kappaB-Inducing Kinase
13.
J Biol Chem ; 291(2): 989-97, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26586915

ABSTRACT

To translate the 13 mtDNA-encoded mRNAs involved in oxidative phosphorylation (OXPHOS), mammalian mitochondria contain a dedicated set of ribosomes comprising rRNAs encoded by the mitochondrial genome and mitochondrial ribosomal proteins (MRPs) that are encoded by nuclear genes and imported into the matrix. In addition to their role in the ribosome, several MRPs have auxiliary functions or have been implicated in other cellular processes like cell cycle regulation and apoptosis. For example, we have shown that human MRPL12 binds and activates mitochondrial RNA polymerase (POLRMT), and hence has distinct functions in the ribosome and mtDNA transcription. Here we provide concrete evidence that there are two mature forms of mammalian MRPL12 that are generated by a two-step cleavage during import, involving efficient cleavage by mitochondrial processing protease and a second inefficient or regulated cleavage by mitochondrial intermediate protease. We also show that knock-down of MRPL12 by RNAi results in instability of POLRMT, but not other primary mitochondrial transcription components, and a corresponding decrease in mitochondrial transcription rates. Knock-down of MRPL10, the binding partner of MRPL12 in the ribosome, results in selective degradation of the mature long form of MRPL12, but has no effect on POLRMT. We propose that the two forms of MRPL12 are involved in homeostatic regulation of mitochondrial transcription and ribosome biogenesis that likely contribute to cell cycle, growth regulation, and longevity pathways to which MRPL12 has been linked.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Mitochondrial Proteins/metabolism , Proteolysis , Ribosomal Proteins/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Mice , Mitochondria/metabolism , Molecular Sequence Data , Protein Isoforms/metabolism , Protein Stability , Protein Transport , Ribosomal Proteins/chemistry , Ribosomes/metabolism , Transcription, Genetic
14.
Mol Cell ; 36(2): 207-18, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19854131

ABSTRACT

Orthologs of the yeast telomere protein Stn1 are present in plants, but other components of the Cdc13/Stn1/Ten1 (CST) complex have only been found in fungi. Here we report the identification of conserved telomere maintenance component 1 (CTC1) in plants and vertebrates. CTC1 encodes an approximately 140 kDa telomere-associated protein predicted to contain multiple OB-fold domains. Arabidopsis mutants null for CTC1 display a severe telomere deprotection phenotype accompanied by a rapid onset of developmental defects and sterility. Telomeric and subtelomeric tracts are dramatically eroded, and chromosome ends exhibit increased G overhangs, recombination, and end-to-end fusions. AtCTC1 both physically and genetically interacts with AtSTN1. Depletion of human CTC1 by RNAi triggers a DNA damage response, chromatin bridges, increased G overhangs, and sporadic telomere loss. These data indicate that CTC1 participates in telomere maintenance in diverse species and that a CST-like complex is required for telomere integrity in multicellular organisms.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Plant/metabolism , Conserved Sequence , Eukaryotic Cells/metabolism , Telomere-Binding Proteins/metabolism , Anaphase , Cell Line, Tumor , Genomic Instability , Humans , In Situ Hybridization, Fluorescence , Mutation/genetics , Nucleic Acid Conformation , Protein Binding , Recombination, Genetic/genetics , Telomere/metabolism
15.
J Am Chem Soc ; 138(11): 3844-55, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-26927829

ABSTRACT

Small-molecule inhibitors of DNA repair pathways are being intensively investigated as primary and adjuvant chemotherapies. We report the discovery that cardiac glycosides, natural products in clinical use for the treatment of heart failure and atrial arrhythmia, are potent inhibitors of DNA double-strand break (DSB) repair. Our data suggest that cardiac glycosides interact with phosphorylated mediator of DNA damage checkpoint protein 1 (phospho-MDC1) or E3 ubiquitin-protein ligase ring finger protein 8 (RNF8), two factors involved in DSB repair, and inhibit the retention of p53 binding protein 1 (53BP1) at the site of DSBs. These observations provide an explanation for the anticancer activity of this class of compounds, which has remained poorly understood for decades, and provide guidance for their clinical applications. This discovery was enabled by the development of the first high-throughput unbiased cellular assay to identify new small-molecule inhibitors of DSB repair. Our assay is based on the fully automated, time-resolved quantification of phospho-SER139-H2AX (γH2AX) and 53BP1 foci, two factors involved in the DNA damage response network, in cells treated with small molecules and ionizing radiation (IR). This primary assay is supplemented by robust secondary assays that establish lead compound potencies and provide further insights into their mechanisms of action. Although the cardiac glycosides were identified in an evaluation of 2366 small molecules, the assay is envisioned to be adaptable to larger compound libraries. The assay is shown to be compatible with small-molecule DNA cleaving agents, such as bleomycin, neocarzinostatin chromophore, and lomaiviticin A, in place of IR.


Subject(s)
Cardiac Glycosides/pharmacology , DNA Breaks, Double-Stranded , DNA End-Joining Repair/drug effects , Fluorescent Antibody Technique/methods , Small Molecule Libraries/pharmacology , Cell Line, Tumor , Humans
16.
Nucleic Acids Res ; 41(4): 2479-88, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23303773

ABSTRACT

Human mitochondrial RNA polymerase, POLRMT, is required for mitochondrial DNA (mtDNA) transcription and forms initiation complexes with human mitochondrial transcription factor B2 (h-mtTFB2). However, POLRMT also interacts with the paralogue of h-mtTFB2, h-mtTFB1, which is a 12S ribosomal RNA methyltransferase required for small (28S) mitochondrial ribosome subunit assembly. Herein, we show that POLRMT associates with h-mtTFB1 in 28S mitochondrial ribosome complexes that are stable in the absence of mitochondrial transcription and distinct from transcription complexes containing POLRMT and h-mtTFB2. Overexpression of POLRMT in HeLa cells increases 12S rRNA methylation by h-mtTFB1 and reduces the steady-state levels of mtDNA-encoded proteins and respiration, apparently because of a decrease in fully assembled 55S mitochondrial ribosomes. We propose that POLRMT interacts directly with h-mtTFB1 in 28S mitochondrial ribosomes to augment its 12S rRNA methyltransferase activity, and that together they provide a checkpoint for proper 28S and 55S mitochondrial ribosome assembly. Thus, POLRMT is multi-functional, forming distinct protein complexes that regulate different steps in mitochondrial gene expression, at least one of which does not involve transcription per se. The significance of these results is discussed with regard to the mechanism and regulation of human mitochondrial gene expression and the potential multi-functionality of RNA polymerases in general.


Subject(s)
DNA-Binding Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Methyltransferases/metabolism , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Ribosomes/enzymology , Transcription Factors/metabolism , Cell Respiration , DNA-Directed RNA Polymerases/physiology , HeLa Cells , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Transcription, Genetic
17.
Proc Natl Acad Sci U S A ; 108(44): 17921-6, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22003127

ABSTRACT

Basal transcription of human mitochondrial DNA (mtDNA) in vitro requires the single-subunit, bacteriophage-related RNA polymerase, POLRMT, and transcription factor h-mtTFB2. This two-component system is activated differentially at mtDNA promoters by human mitochondrial transcription factor A (h-mtTFA). Mitochondrial ribosomal protein L7/L12 (MRPL12) binds directly to POLRMT, but whether it does so in the context of the ribosome or as a "free" protein in the matrix is unknown. Furthermore, existing evidence that MRPL12 activates mitochondrial transcription derives from overexpression studies in cultured cells and transcription experiments using crude mitochondrial lysates, precluding direct effects of MRPL12 on transcription to be assigned. Here, we report that depletion of MRPL12 from HeLa cells by shRNA results in decreased steady-state levels of mitochondrial transcripts, which are not accounted for by changes in RNA stability. We also show that a significant "free" pool of MRPL12 exists in human mitochondria not associated with ribosomes. "Free" MRPL12 binds selectively to POLRMT in vivo in a complex distinct from those containing h-mtTFB2. Finally, using a fully recombinant mitochondrial transcription system, we demonstrate that MRPL12 stimulates promoter-dependent and promoter-independent transcription directly in vitro. Based on these results, we propose that, when not associated with ribosomes, MRPL12 has a second function in transcription, perhaps acting to facilitate the transition from initiation to elongation. We speculate that this is one mechanism to coordinate mitochondrial ribosome biogenesis and transcription in human mitochondria, where transcription of rRNAs from the mtDNA presumably needs to be adjusted in accordance with the rate of import and assembly of the nucleus-encoded MRPs into ribosomes.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Mitochondria/enzymology , Ribosomal Proteins/metabolism , Transcription, Genetic , HeLa Cells , Humans , Real-Time Polymerase Chain Reaction
18.
bioRxiv ; 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-36824951

ABSTRACT

While microRNAs and other non-coding RNAs are the next frontier of novel regulators of mammalian ribosome biogenesis (RB), a systematic exploration of microRNA-mediated RB regulation has not yet been undertaken. We carried out a high-content screen in MCF10A cells for changes in nucleolar number using a library of 2,603 mature human microRNA mimics. Following a secondary screen for nucleolar rRNA biogenesis inhibition, we identified 72 novel microRNA negative regulators of RB after stringent hit calling. Hits included 27 well-conserved microRNAs present in MirGeneDB, and were enriched for mRNA targets encoding proteins with nucleolar localization or functions in cell cycle regulation. Rigorous selection and validation of a subset of 15 microRNA hits unexpectedly revealed that most of them caused dysregulated pre-rRNA processing, elucidating a novel role for microRNAs in RB regulation. Almost all hits impaired global protein synthesis and upregulated CDKN1A ( p21 ) levels, while causing diverse effects on RNA Polymerase 1 (RNAP1) transcription and TP53 protein levels. We discovered that the MIR-28 siblings, hsa-miR-28-5p and hsa-miR-708-5p, directly and potently target the ribosomal protein mRNA RPS28 via tandem primate-specific 3' UTR binding sites, causing a severe pre-18S pre-rRNA processing defect. Our work illuminates novel microRNA attenuators of RB, forging a promising new path for microRNA mimic chemotherapeutics.

19.
Cell Rep Med ; 4(3): 100976, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36921598

ABSTRACT

Restrictive cardiomyopathy (RCM) is defined as increased myocardial stiffness and impaired diastolic relaxation leading to elevated ventricular filling pressures. Human variants in filamin C (FLNC) are linked to a variety of cardiomyopathies, and in this study, we investigate an in-frame deletion (c.7416_7418delGAA, p.Glu2472_Asn2473delinAsp) in a patient with RCM. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with this variant display impaired relaxation and reduced calcium kinetics in 2D culture when compared with a CRISPR-Cas9-corrected isogenic control line. Similarly, mutant engineered cardiac tissues (ECTs) demonstrate increased passive tension and impaired relaxation velocity compared with isogenic controls. High-throughput small-molecule screening identifies phosphodiesterase 3 (PDE3) inhibition by trequinsin as a potential therapy to improve cardiomyocyte relaxation in this genotype. Together, these data demonstrate an engineered cardiac tissue model of RCM and establish the translational potential of this precision medicine approach to identify therapeutics targeting myocardial relaxation.


Subject(s)
Cardiomyopathy, Restrictive , Humans , Cardiomyopathy, Restrictive/genetics , Tissue Engineering , Myocytes, Cardiac , Myocardium , Drug Discovery
20.
J Biol Chem ; 286(48): 41253-41264, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21971050

ABSTRACT

Impaired oxidative phosphorylation (OXPHOS) is implicated in several metabolic disorders. Even though mitochondrial DNA encodes several subunits critical for OXPHOS, the metabolic consequence of activating mitochondrial transcription remains unclear. We show here that LRP130, a protein involved in Leigh syndrome, increases hepatic ß-fatty acid oxidation. Using convergent genetic and biochemical approaches, we demonstrate LRP130 complexes with the mitochondrial RNA polymerase to activate mitochondrial transcription. Activation of mitochondrial transcription is associated with increased OXPHOS activity, increased supercomplexes, and denser cristae, independent of mitochondrial biogenesis. Consistent with increased oxidative phosphorylation, ATP levels are increased in both cells and mouse liver, whereas coupled respiration is increased in cells. We propose activation of mitochondrial transcription remodels mitochondria and enhances oxidative metabolism.


Subject(s)
Fatty Acids/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Oxidative Phosphorylation , Animals , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Fatty Acids/genetics , Hep G2 Cells , Humans , Leigh Disease/genetics , Leigh Disease/metabolism , Mice , Mitochondria, Liver/genetics , Mitochondrial Proteins/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neoplasm Proteins/genetics , Oxidation-Reduction , Oxygen Consumption/physiology , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL