Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Theranostics ; 9(24): 7268-7281, 2019.
Article in English | MEDLINE | ID: mdl-31695767

ABSTRACT

Rationale: An imbalance between protein synthesis and degradation is one of the mechanisms of cardiac hypertrophy. Increased transcription in cardiomyocytes can lead to excessive protein synthesis and cardiac hypertrophy. Maf1 is an RNA polymerase III (RNA pol III) inhibitor that plays a pivotal role in regulating transcription. However, whether Maf1 regulates of cardiac hypertrophy remains unclear. Methods: Cardiac hypertrophy was induced in vivo by thoracic aortic banding (AB) surgery. Both the in vivo and in vitro gain- and loss-of-function experiments by Maf1 knockout (KO) mice and adenoviral transfection were used to verify the role of Maf1 in cardiac hypertrophy. RNA pol III and ERK1/2 inhibitor were utilized to identify the effects of RNA pol III and ERK1/2. The possible interaction between Maf1 and ERK1/2 was clarified by immunoprecipitation (IP) analysis. Results: Four weeks after surgery, Maf1 KO mice exhibited significantly exacerbated AB-induced cardiac hypertrophy characterized by increased heart size, cardiomyocyte surface area, and atrial natriuretic peptide (ANP) expression and by exacerbated pulmonary edema. Also, the deficiency of Maf1 causes more severe cardiac dilation and dysfunction than wild type (WT) mice after pressure overload. In contrast, compared with adenoviral-GFP injected mice, mice injected with adenoviral-Maf1 showed significantly ameliorated AB-induced cardiac hypertrophy. In vitro study has demonstrated that Maf1 could significantly block phenylephrine (PE)-induced cardiomyocyte hypertrophy by inhibiting RNA pol III transcription. However, application of an RNA pol III inhibitor markedly improved Maf1 knockdown-promoted cardiac hypertrophy. Moreover, ERK1/2 was identified as a regulator of RNA pol III, and ERK1/2 inhibition by U0126 significantly repressed Maf1 knockdown-promoted cardiac hypertrophy accompanied by suppressed RNA pol III transcription. Additionally, IP analysis demonstrated that Maf1 could directly bind ERK1/2, suggesting Maf1 could interact with ERK1/2 and then inhibit RNA pol III transcription so as to attenuate the development of cardiac hypertrophy. Conclusions: Maf1 ameliorates PE- and AB-induced cardiac hypertrophy by inhibiting RNA pol III transcription via ERK1/2 signaling suppression.


Subject(s)
Cardiomegaly/metabolism , RNA Polymerase III/metabolism , Repressor Proteins/metabolism , Animals , Cardiomegaly/etiology , Cardiomegaly/genetics , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylephrine/adverse effects , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/genetics , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics
2.
Mol Cell Biol ; 40(1)2019 12 11.
Article in English | MEDLINE | ID: mdl-31658995

ABSTRACT

Mycophenolic acid (MPA) is the active metabolite of mycophenolate mofetil, a drug that is widely used for immunosuppression in organ transplantation and autoimmune diseases, as well as anticancer chemotherapy. It inhibits IMP dehydrogenase, a rate-limiting enzyme in de novo synthesis of guanidine nucleotides. MPA treatment interferes with transcription elongation, resulting in a drastic reduction of pre-rRNA and pre-tRNA synthesis, the disruption of the nucleolus, and consequently cell cycle arrest. Here, we investigated the mechanism whereby MPA inhibits RNA polymerase III (Pol III) activity, in both yeast and mammalian cells. We show that MPA rapidly inhibits Pol III by depleting GTP. Although MPA treatment can activate p53, this is not required for Pol III transcriptional inhibition. The Pol III repressor MAF1 is also not responsible for inhibiting Pol III in response to MPA treatment. We show that upon MPA treatment, the levels of selected Pol III subunits decrease, but this is secondary to transcriptional inhibition. Chromatin immunoprecipitation (ChIP) experiments show that Pol III does not fully dissociate from tRNA genes in yeast treated with MPA, even though there is a sharp decrease in the levels of newly transcribed tRNAs. We propose that in yeast, GTP depletion may lead to Pol III stalling.


Subject(s)
Enzyme Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/pharmacology , RNA Polymerase III/antagonists & inhibitors , RNA, Transfer/genetics , Transcription, Genetic/drug effects , Animals , Gene Expression Regulation, Fungal/drug effects , Mice , RAW 264.7 Cells , RNA Polymerase III/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/metabolism
3.
Nucleic Acids Res ; 47(8): 3937-3956, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30820548

ABSTRACT

RNA polymerase (pol) III occurs in two forms, containing either the POLR3G subunit or the related paralogue POLR3GL. Whereas POLR3GL is ubiquitous, POLR3G is enriched in undifferentiated cells. Depletion of POLR3G selectively triggers proliferative arrest and differentiation of prostate cancer cells, responses not elicited when POLR3GL is depleted. A small molecule pol III inhibitor can cause POLR3G depletion, induce similar differentiation and suppress proliferation and viability of cancer cells. This response involves control of the fate-determining factor NANOG by small RNAs derived from Alu short interspersed nuclear elements. Tumour initiating activity in vivo can be reduced by transient exposure to the pol III inhibitor. Untransformed prostate cells appear less sensitive than cancer cells to pol III depletion or inhibition, raising the possibility of a therapeutic window.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/drug therapy , RNA Polymerase III/genetics , Small Molecule Libraries/pharmacology , Aged , Alu Elements/drug effects , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Chaperone BiP , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Knockout , Middle Aged , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
4.
PLoS Genet ; 14(2): e1007202, 2018 02.
Article in English | MEDLINE | ID: mdl-29401457

ABSTRACT

The small G-protein Ras is a conserved regulator of cell and tissue growth. These effects of Ras are mediated largely through activation of a canonical RAF-MEK-ERK kinase cascade. An important challenge is to identify how this Ras/ERK pathway alters cellular metabolism to drive growth. Here we report on stimulation of RNA polymerase III (Pol III)-mediated tRNA synthesis as a growth effector of Ras/ERK signalling in Drosophila. We find that activation of Ras/ERK signalling promotes tRNA synthesis both in vivo and in cultured Drosophila S2 cells. We also show that Pol III function is required for Ras/ERK signalling to drive proliferation in both epithelial and stem cells in Drosophila tissues. We find that the transcription factor Myc is required but not sufficient for Ras-mediated stimulation of tRNA synthesis. Instead we show that Ras signalling promotes Pol III function and tRNA synthesis by phosphorylating, and inhibiting the nuclear localization and function of the Pol III repressor Maf1. We propose that inhibition of Maf1 and stimulation of tRNA synthesis is one way by which Ras signalling enhances protein synthesis to promote cell and tissue growth.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/genetics , MAP Kinase Signaling System/physiology , RNA, Transfer/biosynthesis , Repressor Proteins/physiology , Transcription Elongation, Genetic , ras Proteins/physiology , Animals , Animals, Genetically Modified , Cell Proliferation/genetics , Cells, Cultured , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Embryo, Nonmammalian , Protein Biosynthesis/genetics , RNA Polymerase III/antagonists & inhibitors , RNA, Transfer/genetics , Repressor Proteins/genetics , Signal Transduction/physiology , Transcription Factor TFIIIB/genetics , Transcription Factor TFIIIB/physiology , Wings, Animal/embryology , Wings, Animal/metabolism
5.
J Virol ; 92(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29142132

ABSTRACT

Despite the availability of two attenuated vaccines, rotavirus (RV) gastroenteritis remains an important cause of mortality among children in developing countries, causing about 215,000 infant deaths annually. Currently, there are no specific antiviral therapies available. RV is a nonenveloped virus with a segmented double-stranded RNA genome. Viral genome replication and assembly of transcriptionally active double-layered particles (DLPs) take place in cytoplasmic viral structures called viroplasms. In this study, we describe strong impairment of the early stages of RV replication induced by a small molecule known as an RNA polymerase III inhibitor, ML-60218 (ML). This compound was found to disrupt already assembled viroplasms and to hamper the formation of new ones without the need for de novo transcription of cellular RNAs. This phenotype was correlated with a reduction in accumulated viral proteins and newly made viral genome segments, disappearance of the hyperphosphorylated isoforms of the viroplasm-resident protein NSP5, and inhibition of infectious progeny virus production. In in vitro transcription assays with purified DLPs, ML showed dose-dependent inhibitory activity, indicating the viral nature of its target. ML was found to interfere with the formation of higher-order structures of VP6, the protein forming the DLP outer layer, without compromising its ability to trimerize. Electron microscopy of ML-treated DLPs showed dose-dependent structural damage. Our data suggest that interactions between VP6 trimers are essential, not only for DLP stability, but also for the structural integrity of viroplasms in infected cells.IMPORTANCE Rotavirus gastroenteritis is responsible for a large number of infant deaths in developing countries. Unfortunately, in the countries where effective vaccines are urgently needed, the efficacy of the available vaccines is particularly low. Therefore, the development of antivirals is an important goal, as they might complement the available vaccines or represent an alternative option. Moreover, they may be decisive in fighting the acute phase of infection. This work describes the inhibitory effect on rotavirus replication of a small molecule initially reported as an RNA polymerase III inhibitor. The molecule is the first chemical compound identified that is able to disrupt viroplasms, the viral replication machinery, and to compromise the stability of DLPs by targeting the viral protein VP6. This molecule thus represents a starting point in the development of more potent and less cytotoxic compounds against rotavirus infection.


Subject(s)
RNA Polymerase III/antagonists & inhibitors , Rotavirus/physiology , Small Molecule Libraries/pharmacology , Viral Structures/drug effects , Animals , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Rotavirus/chemistry , Rotavirus/drug effects , Sf9 Cells , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects
6.
Nature ; 552(7684): 263-267, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29186112

ABSTRACT

Three distinct RNA polymerases transcribe different classes of genes in the eukaryotic nucleus. RNA polymerase (Pol) III is the essential, evolutionarily conserved enzyme that generates short, non-coding RNAs, including tRNAs and 5S rRNA. The historical focus on transcription of protein-coding genes has left the roles of Pol III in organismal physiology relatively unexplored. Target of rapamycin kinase complex 1 (TORC1) regulates Pol III activity, and is also an important determinant of longevity. This raises the possibility that Pol III is involved in ageing. Here we show that Pol III limits lifespan downstream of TORC1. We find that a reduction in Pol III extends chronological lifespan in yeast and organismal lifespan in worms and flies. Inhibiting the activity of Pol III in the gut of adult worms or flies is sufficient to extend lifespan; in flies, longevity can be achieved by Pol III inhibition specifically in intestinal stem cells. The longevity phenotype is associated with amelioration of age-related gut pathology and functional decline, dampened protein synthesis and increased tolerance of proteostatic stress. Pol III acts on lifespan downstream of TORC1, and limiting Pol III activity in the adult gut achieves the full longevity benefit of systemic TORC1 inhibition. Hence, Pol III is a pivotal mediator of this key nutrient-signalling network for longevity; the growth-promoting anabolic activity of Pol III mediates the acceleration of ageing by TORC1. The evolutionary conservation of Pol III affirms its potential as a therapeutic target.


Subject(s)
Longevity/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , RNA Polymerase III/metabolism , Aging/drug effects , Aging/physiology , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/physiology , Drosophila melanogaster/drug effects , Drosophila melanogaster/enzymology , Drosophila melanogaster/physiology , Evolution, Molecular , Female , Food , Intestines/cytology , Intestines/enzymology , Longevity/drug effects , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Protein Biosynthesis , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/deficiency , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/physiology , Stem Cells/cytology , Stem Cells/enzymology
7.
Sci Rep ; 7: 45460, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28368037

ABSTRACT

We carried out padlock capture, a high-resolution RNA allelotyping method, to study X chromosome inactivation (XCI). We examined the gene reactivation pattern along the inactive X (Xi), after Xist (X-inactive specific transcript), a prototype long non-coding RNA essential for establishing X chromosome inactivation (XCI) in early embryos, is conditionally deleted from Xi in somatic cells (Xi∆Xist). We also monitored the behaviors of X-linked non-coding transcripts before and after XCI. In each mutant cell line, gene reactivation occurs to ~6% genes along Xi∆Xist in a recognizable pattern. Genes with upstream regions enriched for SINEs are prone to be reactivated. SINE is a class of retrotransposon transcribed by RNA polymerase III (Pol III). Intriguingly, a significant fraction of Pol III transcription from non-coding regions is not subjected to Xist-mediated transcriptional silencing. Pol III inhibition affects gene reactivation status along Xi∆Xist, alters chromatin configuration and interferes with the establishment XCI during in vitro differentiation of ES cells. These results suggest that Pol III transcription is involved in chromatin structure re-organization during the onset of XCI and functions as a general mechanism regulating chromatin configuration in mammalian cells.


Subject(s)
Chromatin/metabolism , RNA Polymerase III/metabolism , X Chromosome/genetics , Alleles , Animals , Cell Line , Databases, Genetic , Genes, X-Linked , Mice , Polymorphism, Single Nucleotide , RNA Interference , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Small Interfering/metabolism , Transcription Factors, TFIII/antagonists & inhibitors , Transcription Factors, TFIII/genetics , Transcription Factors, TFIII/metabolism , X Chromosome/metabolism
8.
FEBS J ; 283(15): 2811-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27059519

ABSTRACT

Electron cryomicroscopy reconstructions of elongating RNA polymerase (Pol) III at 3.9 Å resolution and of unbound Pol III (apo Pol III) in two distinct conformations at 4.6 Å and 4.7 Å resolution allow the construction of complete atomic models of Pol III and provide new functional insights into the adaption of Pol III to fulfill its specific transcription tasks.


Subject(s)
RNA Polymerase III/chemistry , Transcription, Genetic , Cryoelectron Microscopy , Humans , Models, Molecular , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/metabolism , RNA Polymerase III/ultrastructure
9.
Insect Mol Biol ; 23(3): 367-80, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24580894

ABSTRACT

Although there are alternative telomere structures, most telomeres contain DNA arrays of short repeats (6-26 bp) maintained by telomerase. Like other diptera, Chironomus riparius has noncanonical telomeres and three subfamilies, TsA, TsB and TsC, of longer sequences (176 bp) are found at their chromosomal ends. Reverse transcription PCR was used to show that different RNAs are transcribed from these sequences. Only one strand from TsA sequences seems to render a noncoding RNA (named CriTER-A); transcripts from both TsB strands were found (CriTER-B and αCriTER-B) but no TsC transcripts were detected. Interestingly, these sequences showed a differential transcriptional response upon heat shock, and they were also differentially affected by inhibitors of RNA polymerase II and RNA polymerase III. A computer search for transcription factor binding sites revealed putative regulatory cis-elements within the transcribed sequence, reinforcing the experimental evidence which suggests that the telomeric repeat might function as a promoter. This work describes the telomeric transcriptome of an insect with non-telomerase telomeres, confirming the evolutionary conservation of telomere transcription. Our data reveal differences in the regulation of telomeric transcripts between control and stressful environmental conditions, supporting the idea that telomeric RNAs could have a relevant role in cellular metabolism in insect cells.


Subject(s)
Chironomidae/genetics , Telomere/genetics , Transcriptome , Animals , Heat-Shock Response , Larva , RNA , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase III/antagonists & inhibitors , RNA, Untranslated
10.
Plant Physiol ; 163(1): 232-42, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23898043

ABSTRACT

Transcription activator-like (TAL) effectors from Xanthomonas species pathogens act as transcription factors in plant cells; however, how TAL effectors activate host transcription is unknown. We found previously that TAL effectors of the citrus canker pathogen Xanthomonas citri, known as PthAs, bind the carboxyl-terminal domain of the sweet orange (Citrus sinensis) RNA polymerase II (Pol II) and inhibit the activity of CsCYP, a cyclophilin associated with the carboxyl-terminal domain of the citrus RNA Pol II that functions as a negative regulator of cell growth. Here, we show that PthA4 specifically interacted with the sweet orange MAF1 (CsMAF1) protein, an RNA polymerase III (Pol III) repressor that controls ribosome biogenesis and cell growth in yeast (Saccharomyces cerevisiae) and human. CsMAF1 bound the human RNA Pol III and rescued the yeast maf1 mutant by repressing tRNA(His) transcription. The expression of PthA4 in the maf1 mutant slightly restored tRNA(His) synthesis, indicating that PthA4 counteracts CsMAF1 activity. In addition, we show that sweet orange RNA interference plants with reduced CsMAF1 levels displayed a dramatic increase in tRNA transcription and a marked phenotype of cell proliferation during canker formation. Conversely, CsMAF1 overexpression was detrimental to seedling growth, inhibited tRNA synthesis, and attenuated canker development. Furthermore, we found that PthA4 is required to elicit cankers in sweet orange leaves and that depletion of CsMAF1 in X. citri-infected tissues correlates with the development of hyperplastic lesions and the presence of PthA4. Considering that CsMAF1 and CsCYP function as canker suppressors in sweet orange, our data indicate that TAL effectors from X. citri target negative regulators of RNA Pol II and Pol III to coordinately increase the transcription of host genes involved in ribosome biogenesis and cell proliferation.


Subject(s)
Citrus/physiology , Plant Diseases/genetics , Plant Proteins/physiology , RNA Polymerase III/antagonists & inhibitors , Xanthomonas , Amino Acid Sequence , Citrus/genetics , Citrus/microbiology , Conserved Sequence , Humans , Molecular Sequence Data , Phylogeny , Plant Proteins/genetics , Plant Proteins/metabolism , Repressor Proteins/chemistry , Saccharomyces cerevisiae/genetics , Sequence Alignment
11.
RNA ; 18(10): 1823-32, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22919049

ABSTRACT

tRNA precursors, which are transcribed by RNA polymerase III, undergo end-maturation, splicing, and base modifications. Hypomodified tRNAs, such as tRNA(Val(AAC)), lacking 7-methylguanosine and 5-methylcytidine modifications, are subject to degradation by a rapid tRNA decay pathway. Here we searched for genes which, when overexpressed, restored stability of tRNA(Val(AAC)) molecules in a modification-deficient trm4Δtrm8Δ mutant. We identified TEF1 and VAS1, encoding elongation factor eEF1A and valyl-tRNA synthetase respectively, which likely protect hypomodified tRNA(Val(AAC)) by direct interactions. We also identified MAF1 whose product is a general negative regulator of RNA polymerase III. Expression of a Maf1-7A mutant that constitutively repressed RNA polymerase III transcription resulted in increased stability of hypomodified tRNA(Val(AAC)). Strikingly, inhibition of tRNA transcription in a Maf1-independent manner, either by point mutation in RNA polymerase III subunit Rpc128 or decreased expression of Rpc17 subunit, also suppressed the turnover of the hypomodified tRNA(Val(AAC)). These results support a model where inhibition of tRNA transcription leads to stabilization of hypomodified tRNA(Val(AAC)) due to more efficient protection by tRNA-interacting proteins.


Subject(s)
RNA Polymerase III/antagonists & inhibitors , RNA Stability/genetics , RNA, Transfer/metabolism , Saccharomyces cerevisiae Proteins/physiology , Transcription Factors/physiology , Transcription, Genetic , Down-Regulation/genetics , Gene Expression Regulation, Fungal , Gene Library , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/physiology , Models, Biological , Mutant Proteins/physiology , Organisms, Genetically Modified , Plasmids/genetics , RNA Polymerase III/metabolism , RNA Polymerase III/physiology , RNA Processing, Post-Transcriptional/genetics , RNA Processing, Post-Transcriptional/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics , Transfection
12.
J Virol ; 86(8): 4527-37, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22345436

ABSTRACT

We have used the RAW 264.7 murine macrophage-like cell line as a platform to characterize the recognition and early signaling response to recombinant adenoviral vectors (rAdV). Infection of RAW 264.7 cells triggers an early response (2 to 6 h postinfection) that includes phosphorylation of the interferon (IFN) response factor 3 (IRF3) transcription factor, upregulation of IRF3 primary response genes (interferon-stimulated gene 56 [ISG56], beta IFN [IFN-ß]), and subsequent type I IFN secondary signaling (STAT1/2 phosphorylation). Using short hairpin RNA (shRNA) lentiviral vectors, we show an essential role for Tank binding kinase 1 (TBK1) in this pathway. Data also support a role for STING (MITA) as an adaptor functioning in response to rAdV infection. Using UV/psoralen (Ps)-inactivated virus to block viral transcription, Ps-inactivated virus stimulated primary (IRF3) and secondary (STAT1/2) activation events to the same degree as untreated virus. IRF3 phosphorylation was not blocked in RAW 264.7 cells pretreated with the RNA polymerase III inhibitor ML60218. However, they were compromised in the type I IFN-dependent secondary response (phosphorylation of STAT1/STAT2). At 24 h postinfection, ML60218-treated cells were compromised in the overall antiviral response. Therefore, initial sensing of rAdV or viral DNA (vDNA) does not depend on viral template transcription, but ML60218 treatment influences cellular cascades required for an antiviral response to rAdV. Using overexpression or knockdown assays, we examined how four DNA sensors influence the antiviral response. Knockdown of DNA Activator of Interferon (DAI) and p204, the murine ortholog to IFI16, had minimal influence on IRF3 phosphorylation. However, knockdown of absent in melanoma 2 (AIM2) and the helicase DDX41 resulted in diminished levels of (pser388)IRF3 following rAdV infection. Based on these data, multiple DNA sensors contribute to an antiviral DNA recognition response, leading to TBK1-dependent IRF3 phosphorylation in RAW 264.7 cells.


Subject(s)
Adenoviridae/physiology , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Macrophages/virology , Adenoviridae/genetics , Adenoviridae/metabolism , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Animals , Cell Line , DEAD-box RNA Helicases , DNA Helicases/metabolism , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Gene Silencing , Genome, Viral , Glycoproteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Models, Biological , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Polymerase III/antagonists & inhibitors , RNA, Small Interfering/metabolism , RNA-Binding Proteins , Signal Transduction/drug effects , Transcription, Genetic
13.
Biochem Biophys Res Commun ; 418(1): 149-55, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22244871

ABSTRACT

The stress response, which can trigger various physiological phenomena, is important for living organisms. For instance, a number of stress-induced granules such as P-body and stress granule have been identified. These granules are formed in the cytoplasm under stress conditions and are associated with translational inhibition and mRNA decay. In the nucleus, there is a focus named nuclear stress body (nSB) that distinguishes these structures from cytoplasmic stress granules. Many splicing factors and long non-coding RNA species localize in nSBs as a result of stress. Indeed, tRNAs respond to several kinds of stress such as heat, oxidation or starvation. Although nuclear accumulation of tRNAs occurs in starved Saccharomyces cerevisiae, this phenomenon is not found in mammalian cells. We observed that initiator tRNA(Met) (Meti) is actively translocated into the nucleus of human cells under heat stress. During this study, we identified unique granules of Meti that overlapped with nSBs. Similarly, elongator tRNA(Met) was translocated into the nucleus and formed granules during heat stress. Formation of tRNA granules is closely related to the translocation ratio. Then, all tRNAs may form the specific granules.


Subject(s)
Cell Nucleus/metabolism , Cytoplasmic Granules/metabolism , Heat-Shock Response , RNA, Transfer, Met/metabolism , Cytoplasmic Granules/drug effects , HeLa Cells , Hot Temperature , Humans , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/metabolism , Sirolimus/pharmacology
14.
Invest New Drugs ; 30(1): 316-26, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20839029

ABSTRACT

BACKGROUND: TAS-106 is a novel nucleoside analog that inhibits RNA polymerases I, II and II and has demonstrated robust antitumor activity in a wide range of models of human cancer in preclinical studies. This study was performed to principally evaluate the feasibility of administering TAS-106 as a bolus intravenous (IV) infusion every 3 weeks. PATIENTS AND METHODS: Patients with advanced solid malignancies were treated with escalating doses of TAS-106 as a single bolus IV infusion every 3 weeks. Plasma and urine sampling were performed during the first course to characterize the pharmacokinetic profile of TAS-106 and assess pharmacodynamic relationships. RESULTS: Thirty patients were treated with 66 courses of TAS-106 at eight dose levels ranging from 0.67-9.46 mg/m(2). A cumulative sensory peripheral neuropathy was the principal dose-limiting toxicity (DLT) of TAS-106 at the 6.31 mg/m(2) dose level, which was determined to be the maximum tolerated dose (MTD). Other mild-moderate drug-related toxicities include asthenia, anorexia, nausea, vomiting, myelosuppression, and dermatologic effects. Major objective antitumor responses were not observed. The pharmacokinetics of TAS-106 were dose-proportional. The terminal elimination half-life (t(1/2)) averaged 11.3 ± 3.3 h. Approximately 71% of TAS-106 was excreted in the urine as unchanged drug. Pharmacodynamic relationships were observed between neuropathy and: C(5min;) AUC(0-inf;) and dermatologic toxicity. CONCLUSIONS: The recommended phase II dose of TAS-106 is 4.21 mg/m(2). However, due to a cumulative drug-related peripheral sensory neuropathy that proved to be dose-limiting, further evaluation of this bolus every 21 day infusion schedule will not be pursued and instead, an alternate dosing schedule of TAS-106 administered as a continuous 24-hour infusion will be explored to decrease C(max) in efforts to minimize peripheral neuropathy and maximize antitumor activity.


Subject(s)
Antineoplastic Agents/administration & dosage , Cytidine/analogs & derivatives , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Neoplasms/drug therapy , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase I/antagonists & inhibitors , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Biotransformation , Cytidine/administration & dosage , Cytidine/adverse effects , Cytidine/pharmacokinetics , Drug Administration Schedule , Enzyme Inhibitors/adverse effects , Feasibility Studies , Female , Half-Life , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Male , Maximum Tolerated Dose , Metabolic Clearance Rate , Middle Aged , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , RNA Polymerase I/metabolism , RNA Polymerase II/metabolism , RNA Polymerase III/metabolism , Texas , Treatment Outcome
16.
Cell ; 143(1): 59-70, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20887893

ABSTRACT

RNA polymerase III (Pol III) transcribes short RNAs required for cell growth. Under stress conditions, the conserved protein Maf1 rapidly represses Pol III transcription. We report the crystal structure of Maf1 and cryo-electron microscopic structures of Pol III, an active Pol III-DNA-RNA complex, and a repressive Pol III-Maf1 complex. Binding of DNA and RNA causes ordering of the Pol III-specific subcomplex C82/34/31 that is required for transcription initiation. Maf1 binds the Pol III clamp and rearranges C82/34/31 at the rim of the active center cleft. This impairs recruitment of Pol III to a complex of promoter DNA with the initiation factors Brf1 and TBP and thus prevents closed complex formation. Maf1 does however not impair binding of a DNA-RNA scaffold and RNA synthesis. These results explain how Maf1 specifically represses transcription initiation from Pol III promoters and indicate that Maf1 also prevents reinitiation by binding Pol III during transcription elongation.


Subject(s)
RNA Polymerase III/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Promoter Regions, Genetic , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/chemistry , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Transcription Factors/chemistry , Transcription, Genetic
17.
J Gen Virol ; 91(Pt 9): 2221-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20519457

ABSTRACT

The vaccinia virus E3 protein is an important intracellular modulator of innate immunity that can be split into distinct halves. The C terminus contains a well defined dsRNA-binding domain, whereas the N terminus contains a Z-DNA-binding domain, and both domains are required for virulence. In this study, we investigated whether the E3 Z-DNA-binding domain functions by sequestering cytoplasmic dsDNA thereby preventing the induction of type I interferon (IFN). In line with this hypothesis, expression of E3 ablated both IFN-beta expression and NF-kappaB activity in response to the dsDNA, poly(dA-dT). However, surprisingly, the ability of E3 to block poly(dA-dT) signalling was independent of the N terminus, whereas the dsRNA-binding domain was essential, suggesting that the Z-DNA-binding domain does not bind immunostimulatory dsDNA. This was confirmed by the failure of E3 to co-precipitate with biotinylated dsDNA, whereas the recruitment of several cytoplasmic DNA-binding proteins could be detected. Recently, AT-rich dsDNA was reported to be transcribed into 5'-triphosphate poly(A-U) RNA by RNA polymerase III, which then activates retinoic acid-inducible gene I (RIG-I). Consistent with this, RNA from poly(dA-dT) transfected cells induced IFN-beta and expression of the E3 dsRNA-binding domain was sufficient to ablate this response. Given the well documented function of the E3 dsRNA-binding domain we propose that E3 blocks signalling in response to poly(dA-dT) by binding to transcribed poly(A-U) RNA preventing RIG-I activation. This report describes a DNA virus-encoded inhibitor of the RNA polymerase III-dsDNA-sensing pathway and extends our knowledge of E3 as a modulator of innate immunity.


Subject(s)
Immunity, Innate , RNA Polymerase III/antagonists & inhibitors , RNA-Binding Proteins/immunology , Vaccinia virus/immunology , Viral Proteins/immunology , Cell Line , DNA, Z-Form/metabolism , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Interferon-beta/biosynthesis , NF-kappa B/metabolism , Poly dA-dT/genetics , Poly dA-dT/metabolism , Protein Structure, Tertiary , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Toll-Like Receptor 9/metabolism , Transfection , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Vaccinia virus/physiology , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
18.
Mol Cell Biol ; 30(15): 3749-57, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20516213

ABSTRACT

mTORC1 is a central regulator of growth in response to nutrient availability, but few direct targets have been identified. RNA polymerase (pol) III produces a number of essential RNA molecules involved in protein synthesis, RNA maturation, and other processes. Its activity is highly regulated, and deregulation can lead to cell transformation. The human phosphoprotein MAF1 becomes dephosphorylated and represses pol III transcription after various stresses, but neither the significance of the phosphorylations nor the kinase involved is known. We find that human MAF1 is absolutely required for pol III repression in response to serum starvation or TORC1 inhibition by rapamycin or Torin1. The protein is phosphorylated mainly on residues S60, S68, and S75, and this inhibits its pol III repression function. The responsible kinase is mTORC1, which phosphorylates MAF1 directly. Our results describe molecular mechanisms by which mTORC1 controls human MAF1, a key repressor of RNA polymerase III transcription, and add a new branch to the signal transduction cascade immediately downstream of TORC1.


Subject(s)
RNA Polymerase III/genetics , RNA Polymerase III/metabolism , Humans , Phosphorylation , RNA Polymerase III/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/metabolism , Sirolimus/pharmacology , Transfection
19.
J Mol Biol ; 387(3): 523-31, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19361418

ABSTRACT

RNA polymerase III (RNA pol III) transcribes structural RNAs involved in RNA processing (U6 snRNA) and translation (tRNA), thereby regulating the growth rate of cells. Proper initiation by RNA pol III requires the transcription factor TFIIIB. Gene-external U6 snRNA transcription requires TFIIIB consisting of Bdp1, TBP, and Brf2. Transcription from the gene-internal tRNA promoter requires TFIIIB composed of Bdp1, TBP, and Brf1. TFIIIB is a target of tumor suppressors, including PTEN, ARF, p53, and RB, and RB-related pocket proteins. Breast cancer susceptibility gene 1 (BRCA1) tumor suppressor plays a role in DNA repair, cell cycle regulation, apoptosis, genome integrity, and ubiquitination. BRCA1 has a conserved amino-terminal RING domain, an activation domain 1 (AD1), and an acidic carboxyl-terminal domain (BRCA1 C-terminal region). In Saccharomyces cerevisiae, TFIIB interacts with the BRCA1 C-terminal region domain of Fcp1p, an RNA polymerase II phosphatase. The TFIIIB subunits Brf1 and Brf2 are structurally similar to TFIIB. Hence, we hypothesize that RNA pol III may be regulated by BRCA1 via the TFIIB family members Brf1 and Brf2. Here we report that: (1) BRCA1 inhibits both VAI (tRNA) and U6 snRNA RNA pol III transcription; (2) the AD1 of BRCA1 is responsible for inhibition of U6 snRNA transcription, whereas the RING domain and AD1 of BRCA1 are required for VAI transcription inhibition; and (3) overexpression of Brf1 and Brf2 alleviates inhibition of U6 snRNA and VAI transcription by BRCA1. Taken together, these data suggest that BRCA1 is a general repressor of RNA pol III transcription.


Subject(s)
BRCA1 Protein/metabolism , RNA Polymerase III/antagonists & inhibitors , RNA Polymerase III/metabolism , Transcription Factor TFIIIB/metabolism , Transcription, Genetic , Aged , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Female , Humans , RNA Polymerase III/genetics , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIIIB/genetics
20.
Nucleic Acids Res ; 36(11): 3757-64, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18487626

ABSTRACT

RNA polymerase (pol) III contains a dissociable subcomplex that is required for initiation, but not for elongation or termination of transcription. This subcomplex is composed of subunits RPC3, RPC6 and RPC7, and interacts with TFIIIB, a factor that is necessary and sufficient to support accurate pol III transcription in vitro. Direct binding of TFIIIB to RPC6 is believed to recruit pol III to its genetic templates. However, this has never been tested in vivo. Here we combine chromatin immunoprecipitation with RNA interference to demonstrate that the RPC3/6/7 subcomplex is required for pol III recruitment in mammalian cells. Specific knockdown of RPC6 by RNAi results in post-transcriptional depletion of the other components of the subcomplex, RPC3 and RPC7, without destabilizing core pol III subunits or TFIIIB. The resultant core enzyme is defective in associating with TFIIIB and target genes in vivo. Promoter occupancy by pol II is unaffected, despite sharing five subunits with the pol III core. These observations provide evidence for the validity in vivo of the model for pol III recruitment that was built on biochemical data.


Subject(s)
Carrier Proteins/metabolism , Protein Subunits/metabolism , RNA Polymerase III/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line , Mice , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , RNA Interference , RNA Polymerase III/antagonists & inhibitors , Transcription Factor TFIIIB/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...