Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 439
Filter
1.
Nucleic Acids Res ; 49(6): 3185-3203, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33693809

ABSTRACT

Protein methylation occurs primarily on lysine and arginine, but also on some other residues, such as histidine. METTL18 is the last uncharacterized member of a group of human methyltransferases (MTases) that mainly exert lysine methylation, and here we set out to elucidate its function. We found METTL18 to be a nuclear protein that contains a functional nuclear localization signal and accumulates in nucleoli. Recombinant METTL18 methylated a single protein in nuclear extracts and in isolated ribosomes from METTL18 knockout (KO) cells, identified as 60S ribosomal protein L3 (RPL3). We also performed an RPL3 interactomics screen and identified METTL18 as the most significantly enriched MTase. We found that His-245 in RPL3 carries a 3-methylhistidine (3MH; τ-methylhistidine) modification, which was absent in METTL18 KO cells. In addition, both recombinant and endogenous METTL18 were found to be automethylated at His-154, thus further corroborating METTL18 as a histidine-specific MTase. Finally, METTL18 KO cells displayed altered pre-rRNA processing, decreased polysome formation and codon-specific changes in mRNA translation, indicating that METTL18-mediated methylation of RPL3 is important for optimal ribosome biogenesis and function. In conclusion, we have here established METTL18 as the second human histidine-specific protein MTase, and demonstrated its functional relevance.


Subject(s)
Protein Biosynthesis , Protein Methyltransferases/metabolism , RNA, Ribosomal/metabolism , Ribosomal Proteins/metabolism , Amino Acid Motifs , Cell Nucleolus/enzymology , HEK293 Cells , HeLa Cells , Histidine/metabolism , Humans , Nuclear Localization Signals , Protein Methyltransferases/chemistry , RNA Processing, Post-Transcriptional , Ribosomal Protein L3 , Ribosomes/metabolism
2.
Cancer Sci ; 112(2): 619-628, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33164285

ABSTRACT

Overexpression of the ubiquitous protein kinase, CK2α, has been reported in various human cancers. Here, we demonstrate that nuclear and nucleolar CK2α localization in invasive ductal carcinomas of the breast is a reliable predictor of poor prognosis. Cellular localization of CK2α in nuclei and nucleoli was analyzed immunohistochemically using surgical tissue blocks from 112 patients, who had undergone surgery without neoadjuvant chemotherapy. Clinical data collection and median follow-up period were for more than 5 y. In total, 93.8% of patients demonstrated elevated CK2α expression in nuclei and 36.6% of them displayed elevated expression predominantly in nucleoli. Clinicopathological malignancy was strongly correlated with elevated nuclear and nucleolar CK2α expression. Recurrence-free survival was significantly worse (P = .0002) in patients with positive nucleolar CK2α staining. The 5-y survival rate decreased to a roughly 50% in nucleolar CK2α-positive patients of triple-negative (P = .0069) and p Stage 3 (P = .0073) groups. In contrast, no patients relapsed or died in the triple-negative group who exhibited a lack of nucleolar CK2α staining. Evaluation of nucleolar CK2α staining showed a high secondary index with a hazard ratio of 6.629 (P = .001), following lymph node metastasis with a hazard ratio of 14.30 (P = .0008). Multivariate analysis demonstrated that nucleolar CK2α is an independent factor for recurrence-free survival. Therefore, we propose that histochemical evaluation of nucleolar CK2α-positive staining may be a new and robust prognostic indicator for patients who need further treatment. Functional consequences of nucleolar CK2 dysfunction may be a starting point to facilitate development of novel treatments for invasive breast carcinoma.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Casein Kinase II/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Cell Nucleolus/enzymology , Cell Nucleus/enzymology , Female , Humans , MCF-7 Cells , Middle Aged , Prognosis
3.
Histol Histopathol ; 35(10): 1181-1187, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32901907

ABSTRACT

Ribosome biogenesis is a fine-tuned cellular process and its deregulation is linked to cancer progression: tumors characterized by an intense ribosome biogenesis often display a more aggressive behavior. Ribosomal RNA (rRNA) synthesis is controlled at several levels, the higher one being the epigenetic regulation of the condensation of chromatin portions containing rRNA genes. KDM2A and KDM2B (Lysine (K)-specific demethylase 2A / B) are histone demethylases modulating the accessibility of ribosomal genes, thereby regulating their transcription. Both enzymes are able to demethylate lysins at relevant sites (e.g. K4, K36) on histone H3. We previously demonstrated that KDM2B is one of the factors regulating ribosome biogenesis in human breast cancer. In this study we aimed to define the combined contribution of KDM2A and KDM2B to breast cancer outcome. KDM2A and KDM2B mRNA levels, nucleolar area as a marker of ribosome biogenesis, and patients' prognosis were retrospectively assessed in a series of primary breast carcinomas. We observed that tumors characterized by reduced levels of both KDM2A and KDM2B displayed a particularly aggressive clinical behavior and increased nucleolar size. Our results suggest that KDM2A and KDM2B may cooperate in regulating ribosome biogenesis thus influencing the biological behavior and clinical outcome of human breast cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Lobular/enzymology , Cell Nucleolus/enzymology , F-Box Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/pathology , Cell Nucleolus/genetics , Cell Nucleolus/pathology , F-Box Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Prognosis , Retrospective Studies , Ribosomes/genetics , Ribosomes/metabolism
4.
Nature ; 585(7824): 298-302, 2020 09.
Article in English | MEDLINE | ID: mdl-32669707

ABSTRACT

Proteins are manufactured by ribosomes-macromolecular complexes of protein and RNA molecules that are assembled within major nuclear compartments called nucleoli1,2. Existing models suggest that RNA polymerases I and III (Pol I and Pol III) are the only enzymes that directly mediate the expression of the ribosomal RNA (rRNA) components of ribosomes. Here we show, however, that RNA polymerase II (Pol II) inside human nucleoli operates near genes encoding rRNAs to drive their expression. Pol II, assisted by the neurodegeneration-associated enzyme senataxin, generates a shield comprising triplex nucleic acid structures known as R-loops at intergenic spacers flanking nucleolar rRNA genes. The shield prevents Pol I from producing sense intergenic noncoding RNAs (sincRNAs) that can disrupt nucleolar organization and rRNA expression. These disruptive sincRNAs can be unleashed by Pol II inhibition, senataxin loss, Ewing sarcoma or locus-associated R-loop repression through an experimental system involving the proteins RNaseH1, eGFP and dCas9 (which we refer to as 'red laser'). We reveal a nucleolar Pol-II-dependent mechanism that drives ribosome biogenesis, identify disease-associated disruption of nucleoli by noncoding RNAs, and establish locus-targeted R-loop modulation. Our findings revise theories of labour division between the major RNA polymerases, and identify nucleolar Pol II as a major factor in protein synthesis and nuclear organization, with potential implications for health and disease.


Subject(s)
Cell Nucleolus/enzymology , Cell Nucleolus/genetics , DNA, Ribosomal/genetics , RNA Polymerase II/metabolism , RNA, Untranslated/biosynthesis , RNA, Untranslated/genetics , Ribosomes/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Cell Line, Tumor , Cell Nucleolus/physiology , DNA Helicases/metabolism , DNA, Intergenic/genetics , Humans , Multifunctional Enzymes/metabolism , Protein Biosynthesis , R-Loop Structures , RNA Helicases/metabolism , RNA Polymerase I/antagonists & inhibitors , RNA Polymerase I/metabolism , Ribonuclease H/metabolism , Ribosomes/chemistry , Ribosomes/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology
5.
Cell Microbiol ; 22(10): e13246, 2020 10.
Article in English | MEDLINE | ID: mdl-32720355

ABSTRACT

Intracellular bacterial pathogens harbour genes, the closest homologues of which are found in eukaryotes. Regulator of chromosome condensation 1 (RCC1) repeat proteins are phylogenetically widespread and implicated in protein-protein interactions, such as the activation of the small GTPase Ran by its cognate guanine nucleotide exchange factor, RCC1. Legionella pneumophila and Coxiella burnetii, the causative agents of Legionnaires' disease and Q fever, respectively, harbour RCC1 repeat coding genes. Legionella pneumophila secretes the RCC1 repeat 'effector' proteins LegG1, PpgA and PieG into eukaryotic host cells, where they promote the activation of the pleiotropic small GTPase Ran, microtubule stabilisation, pathogen vacuole motility and intracellular bacterial growth as well as host cell migration. The RCC1 repeat effectors localise to the pathogen vacuole or the host plasma membrane and target distinct components of the Ran GTPase cycle, including Ran modulators and the small GTPase itself. Coxiella burnetii translocates the RCC1 repeat effector NopA into host cells, where the protein localises to nucleoli. NopA binds to Ran GTPase and promotes the nuclear accumulation of Ran(GTP), thus pertubing the import of the transcription factor NF-κB and innate immune signalling. Hence, divergent evolution of bacterial RCC1 repeat effectors defines the range of Ran GTPase cycle targets and likely allows fine-tuning of Ran GTPase activation by the pathogens at different cellular sites.


Subject(s)
Biological Evolution , Coxiella burnetii/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Legionella pneumophila/metabolism , ran GTP-Binding Protein/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Nucleolus/enzymology , Coxiella burnetii/genetics , Coxiella burnetii/pathogenicity , Enzyme Activation , Genes, Bacterial , Host-Pathogen Interactions , Humans , Legionella/genetics , Legionella/metabolism , Legionella/pathogenicity , Legionella pneumophila/genetics , Legionnaires' Disease/microbiology , Protein Transport , Q Fever/microbiology , Vacuoles/metabolism , Vacuoles/microbiology
6.
Cell Biol Int ; 42(11): 1463-1466, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30080298

ABSTRACT

Fibrillarin is an essential nucleolar protein that catalyzes the 2'-O-methylation of ribosomal RNAs. Recently, experimental data have begun to accumulate that suggest that fibrillarin can influence various cellular processes, development of pathological processes, and even aging. The exact mechanism by which fibrillarin can influence these processes has not been found, but some experimental data indicate that up- or downregulation of fibrillarin can modify the ribosome structure and, thus, causе an alteration in relative efficiency with which various mRNAs are translated. Here, we discuss recent studies on the potential roles of fibrillarin in the regulation of cell proliferation, cancer progression, and aging.


Subject(s)
Aging/metabolism , Cell Nucleolus/enzymology , Chromosomal Proteins, Non-Histone/metabolism , Methyltransferases/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Cell Proliferation , Humans
7.
Nucleic Acids Res ; 46(12): 6304-6317, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29618122

ABSTRACT

Among the proteins predicted to be a part of the DExD box RNA helicase family, the functions of DDX49 are unknown. Here, we characterize the enzymatic activities and functions of DDX49 by comparing its properties with the well-studied RNA helicase, DDX39B. We find that DDX49 exhibits a robust ATPase and RNA helicase activity, significantly higher than that of DDX39B. DDX49 is required for the efficient export of poly (A)+ RNA from nucleus in a splicing-independent manner. Furthermore, DDX49 is a resident protein of nucleolus and regulates the steady state levels of pre-ribosomal RNA by regulating its transcription and stability. These dual functions of regulating mRNA export and pre-ribosomal RNA levels enable DDX49 to modulate global translation. Phenotypically, DDX49 promotes proliferation and colony forming potential of cells. Strikingly, DDX49 is significantly elevated in diverse cancer types suggesting that the increased abundance of DDX49 has a role in oncogenic transformation of cells. Taken together, this study shows the physiological role of DDX49 in regulating distinct steps of mRNA and pre-ribosomal RNA metabolism and hence translation and potential pathological role of its dysregulation, especially in cancers.


Subject(s)
DEAD-box RNA Helicases/metabolism , Protein Biosynthesis , RNA Helicases/metabolism , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA, Ribosomal/metabolism , Adenosine Triphosphate/metabolism , Carcinogenesis , Cell Line , Cell Nucleolus/enzymology , Cell Nucleolus/genetics , Cell Proliferation , DEAD-box RNA Helicases/genetics , Humans , RNA Precursors/biosynthesis , RNA Stability , RNA Transport
8.
Nucleic Acids Res ; 45(18): 10672-10692, 2017 Oct 13.
Article in English | MEDLINE | ID: mdl-28977560

ABSTRACT

An R-loop is a DNA:RNA hybrid formed during transcription when a DNA duplex is invaded by a nascent RNA transcript. R-loops accumulate in nucleoli during RNA polymerase I (RNAP I) transcription. Here, we report that mammalian RNase H1 enriches in nucleoli and co-localizes with R-loops in cultured human cells. Co-migration of RNase H1 and R-loops from nucleoli to perinucleolar ring structures was observed upon inhibition of RNAP I transcription. Treatment with camptothecin which transiently stabilized nucleolar R-loops recruited RNase H1 to the nucleoli. It has been reported that the absence of Topoisomerase and RNase H activity in Escherichia coli or Saccharomyces cerevisiae caused R-loop accumulation along rDNA. We found that the distribution of RNase H1 and Top1 along rDNA coincided at sites where R-loops accumulated in mammalian cells. Loss of either RNase H1 or Top1 caused R-loop accumulation, and the accumulation of R-loops was exacerbated when both proteins were depleted. Importantly, we observed that protein levels of Top1 were negatively correlated with the abundance of RNase H1. We conclude that Top1 and RNase H1 are partially functionally redundant in mammalian cells to suppress RNAP I transcription-associate R-loops.


Subject(s)
Cell Nucleolus/genetics , DNA, Ribosomal/chemistry , RNA Polymerase I/metabolism , Ribonuclease H/analysis , Transcription, Genetic , Animals , Camptothecin/pharmacology , Cell Nucleolus/drug effects , Cell Nucleolus/enzymology , DNA Damage , DNA Topoisomerases, Type I/analysis , DNA, Ribosomal/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice, Knockout , Protein Domains , RNA/chemistry , RNA Polymerase I/analysis , Ribonuclease H/chemistry , Ribonuclease H/metabolism , Transcription, Genetic/drug effects
9.
Nucleus ; 8(4): 421-432, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28622108

ABSTRACT

Nucleoli are formed on the basis of ribosomal DNA (rDNA) clusters called Nucleolus Organizer Regions (NORs). Each NOR contains multiple genes coding for RNAs of the ribosomal particles. The prominent components of the nucleolar ultrastructure, fibrillar centers (FC) and dense fibrillar components (DFC), together compose FC/DFC units. These units are centers of rDNA transcription by RNA polymerase I (pol I), as well as the early processing events, in which an essential role belongs to fibrillarin. Each FC/DFC unit probably corresponds to a single transcriptionally active gene. In this work, we transfected human-derived cells with GFP-RPA43 (subunit of pol I) and RFP-fibrillarin. Following changes of the fluorescent signals in individual FC/DFC units, we found two kinds of kinetics: 1) the rapid fluctuations with periods of 2-3 min, when the pol I and fibrillarin signals oscillated in anti-phase manner, and the intensities of pol I in the neighboring FC/DFC units did not correlate. 2) fluctuations with periods of 10 to 60 min, in which pol I and fibrillarin signals measured in the same unit did not correlate, but pol I signals in the units belonging to different nucleoli were synchronized. Our data indicate that a complex pulsing activity of transcription as well as early processing is common for ribosomal genes.


Subject(s)
Cell Nucleolus/chemistry , Cell Nucleolus/enzymology , Chromosomal Proteins, Non-Histone/metabolism , DNA-Directed RNA Polymerases/metabolism , Chromosomal Proteins, Non-Histone/chemistry , DNA-Directed RNA Polymerases/chemistry , HeLa Cells , Humans , Immunohistochemistry , Microscopy, Confocal
10.
J Cell Biol ; 216(8): 2355-2371, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28637749

ABSTRACT

Telomerase can generate a novel telomere at DNA double-strand breaks (DSBs), an event called de novo telomere addition. How this activity is suppressed remains unclear. Combining single-molecule imaging and deep sequencing, we show that the budding yeast telomerase RNA (TLC1 RNA) is spatially segregated to the nucleolus and excluded from sites of DNA repair in a cell cycle-dependent manner. Although TLC1 RNA accumulates in the nucleoplasm in G1/S, Pif1 activity promotes TLC1 RNA localization in the nucleolus in G2/M. In the presence of DSBs, TLC1 RNA remains nucleolar in most G2/M cells but accumulates in the nucleoplasm and colocalizes with DSBs in rad52Δ cells, leading to de novo telomere additions. Nucleoplasmic accumulation of TLC1 RNA depends on Cdc13 localization at DSBs and on the SUMO ligase Siz1, which is required for de novo telomere addition in rad52Δ cells. This study reveals novel roles for Pif1, Rad52, and Siz1-dependent sumoylation in the spatial exclusion of telomerase from sites of DNA repair.


Subject(s)
Cell Cycle , Cell Nucleolus/enzymology , DNA Breaks, Double-Stranded , DNA Repair , DNA, Fungal/metabolism , RNA, Fungal/metabolism , RNA/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Telomerase/metabolism , Telomere/metabolism , Active Transport, Cell Nucleus , Bleomycin/toxicity , Cell Cycle/drug effects , Cell Nucleolus/drug effects , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair/drug effects , DNA, Fungal/genetics , High-Throughput Nucleotide Sequencing , RNA/genetics , RNA, Fungal/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Single Molecule Imaging , Sumoylation , Telomerase/genetics , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Time Factors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Genes Dev ; 31(8): 719-720, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28512234

ABSTRACT

SUMO homeostasis is important for many cellular processes. In the current issue of Genes & Development, Liang and colleagues (pp. 802-815) demonstrate how a desumoylation enzyme is targeted to the nucleolus for removing SUMO from specific substrates and how curtailing sumoylation levels can regulate transcription in this nuclear compartment.


Subject(s)
Gene Expression Regulation , Homeostasis/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Animals , Cell Nucleolus/enzymology , DNA, Ribosomal/metabolism , Endopeptidases/metabolism , Humans , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/metabolism
12.
J Cell Biol ; 216(6): 1795-1810, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28432080

ABSTRACT

The PIDDosome (PIDD-RAIDD-caspase-2 complex) is considered to be the primary signaling platform for caspase-2 activation in response to genotoxic stress. Yet studies of PIDD-deficient mice show that caspase-2 activation can proceed in the absence of PIDD. Here we show that DNA damage induces the assembly of at least two distinct activation platforms for caspase-2: a cytoplasmic platform that is RAIDD dependent but PIDD independent, and a nucleolar platform that requires both PIDD and RAIDD. Furthermore, the nucleolar phosphoprotein nucleophosmin (NPM1) acts as a scaffold for PIDD and is essential for PIDDosome assembly in the nucleolus after DNA damage. Inhibition of NPM1 impairs caspase-2 processing, apoptosis, and caspase-2-dependent inhibition of cell growth, demonstrating that the NPM1-dependent nucleolar PIDDosome is a key initiator of the caspase-2 activation cascade. Thus we have identified the nucleolus as a novel site for caspase-2 activation and function.


Subject(s)
Apoptosis , Caspase 2/metabolism , Cell Nucleolus/enzymology , Cysteine Endopeptidases/metabolism , DNA Damage , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Nuclear Proteins/metabolism , Animals , CRADD Signaling Adaptor Protein/metabolism , Caspase 2/genetics , Cysteine Endopeptidases/genetics , Death Domain Receptor Signaling Adaptor Proteins/genetics , Enzyme Activation , Genotype , HEK293 Cells , HeLa Cells , Humans , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Video , Multiprotein Complexes , Nuclear Proteins/genetics , Nucleophosmin , Phenotype , Protein Binding , RNA Interference , Signal Transduction , Transfection , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
J Cell Biochem ; 118(2): 407-419, 2017 02.
Article in English | MEDLINE | ID: mdl-27428351

ABSTRACT

Human topoisomerase I is partitioned between the nucleolus and the nucleoplasm in the interphase cells. Under unstressed conditions it is concentrated in the first compartment but nucleolar concentration of the full length protein is lost after inactivation of relaxation activity. Due to the above, subnuclear localization of topoisomerase I is linked with DNA relaxation activity of topoisomerase I. Looking for other factors responsible for subnuclear distribution of topoisomerase I, we studied here localization of the fluorescently tagged fragments and point mutants of topoisomerase I in HeLa cells. We found that two regions of topoisomerase I, the N-terminal and the linker domains, were critical for subnuclear localization of the enzyme. The linker domain and the distal region of the N-terminal domain directed topoisomerase I to the nucleolus, whereas the remaining region of the N-terminal domain was responsible for the nucleoplasmic localization. The effects exhibited by the regions which contributed to nuclear distribution of topoisomerase I were independent of DNA relaxation activity. Localization mutations in both domains complemented one another giving the wild-type phenotype for the double mutant. These results suggest a two-stage model of regulation of partitioning of topoisomerase I between the nucleolus and the nucleoplasm. The first stage is a net of interactions provided by the N-terminal and the linker domains. The other stage, accessible only if the first net is balanced, is driven by DNA relaxation activity. J. Cell. Biochem. 118: 407-419, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Nucleolus/enzymology , DNA Topoisomerases, Type I/metabolism , Cell Nucleolus/genetics , DNA Topoisomerases, Type I/genetics , HeLa Cells , Humans , Microscopy, Fluorescence , Recombinant Fusion Proteins
14.
Semin Cell Dev Biol ; 63: 154-166, 2017 03.
Article in English | MEDLINE | ID: mdl-27867042

ABSTRACT

Neurodegeneration is a salient feature of chronic refractory brain disorders like Alzheimer's, Parkinson's, Huntington's, amyotropic lateral sclerosis and acute conditions like cerebral ischemia/reperfusion etc. The pathological protein aggregates, mitochondrial mutations or ischemic insults typifying these disease conditions collude with and intensify existing oxidative stress and attendant mitochondrial dysfunction. Interlocking these mechanisms is poly(ADP-ribose) polymerase (PARP-1) hyperactivation that invokes a distinct form of neuronal cell death viz., 'parthanatos'. PARP-1, a typical 'moonlighting protein' by virtue of its ability to poly(ADP-ribosyl)ate a plethora of cellular proteins exerts diverse functions that impinge significantly on cellular processes. In addition, its interactions with various nuclear proteins like transcription factors and chromatin modifiers elicit varied transcriptional outcomes that wield pathological cellular responses. Further, emerging leitmotifs like mitochondrial and nucleolar PARPs and the novel aspects of gene expression regulation by PARP-1 and poly(ADP-ribosyl)ation can provide a holistic view of PARP-1's influence on cell vitality. In this review, we discuss the pathological underpinnings of PARP-1, with a special emphasis on mitochondrial dysfunction and cell death subroutines, in the realm of neurodegeneration. This would provide a deeper insight into the functions of PARP-1 in neurodegenerative conditions that would enable the development of more effective therapeutic strategies.


Subject(s)
Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Neurons/enzymology , Neurons/pathology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cell Nucleolus/enzymology , Humans , Mitochondria/metabolism , Protein Interaction Maps
15.
Virology ; 500: 169-177, 2017 01.
Article in English | MEDLINE | ID: mdl-27821284

ABSTRACT

Flaviviruses, including the human pathogen, West Nile virus (WNV), are known to co-opt many host factors for their replication and propagation. To this end, we previously reported that the nucleolar DEAD-box RNA helicase, DDX56, is important for production of infectious WNV virions. In this study, we show that WNV infection results in relocalization of DDX56 from nucleoli to virus assembly sites on the endoplasmic reticululm (ER), an observation that is consistent with a role for DDX56 in WNV virion assembly. Super-resolution microscopy revealed that capsid and DDX56 localized to the same subcompartment of the ER, however, unexpectedly, stable interaction between these two proteins was only detected in the nucleus. Together, these data suggest that DDX56 relocalizes to the site of virus assembly during WNV infection and that its interaction with WNV capsid in the cytoplasm may occur transiently during virion morphogenesis.


Subject(s)
Cell Nucleolus/enzymology , DEAD-box RNA Helicases/metabolism , Virus Assembly , West Nile Fever/enzymology , West Nile virus/physiology , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Nucleolus/genetics , Cell Nucleolus/virology , Cytoplasm/metabolism , Cytoplasm/virology , DEAD-box RNA Helicases/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Host-Pathogen Interactions , Humans , Protein Transport , Virus Replication , West Nile Fever/genetics , West Nile Fever/virology , West Nile virus/genetics
16.
PLoS Genet ; 12(2): e1005844, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26848586

ABSTRACT

Plants have evolved a considerable number of intrinsic tolerance strategies to acclimate to ambient temperature increase. However, their molecular mechanisms remain largely obscure. Here we report a DEAD-box RNA helicase, TOGR1 (Thermotolerant Growth Required1), prerequisite for rice growth themotolerance. Regulated by both temperature and the circadian clock, its expression is tightly coupled to daily temperature fluctuations and its helicase activities directly promoted by temperature increase. Located in the nucleolus and associated with the small subunit (SSU) pre-rRNA processome, TOGR1 maintains a normal rRNA homeostasis at high temperature. Natural variation in its transcript level is positively correlated with plant height and its overexpression significantly improves rice growth under hot conditions. Our findings reveal a novel molecular mechanism of RNA helicase as a key chaperone for rRNA homeostasis required for rice thermotolerant growth and provide a potential strategy to breed heat-tolerant crops by modulating the expression of TOGR1 and its orthologs.


Subject(s)
Adaptation, Physiological , Cell Nucleolus/enzymology , DEAD-box RNA Helicases/metabolism , Oryza/physiology , Plant Proteins/metabolism , RNA Precursors/metabolism , Temperature , Cell Proliferation , Circadian Rhythm/genetics , Mutation/genetics , Oryza/cytology , Oryza/enzymology , Plant Development , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/genetics , Ribosome Subunits, Small/metabolism
17.
Cell Signal ; 27(10): 2045-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26215099

ABSTRACT

Increased rates of ribosome biogenesis and biomass accumulation are fundamental properties of rapidly growing and dividing malignant cells. The MYC oncoprotein drives growth predominantly via its ability to upregulate the ribosome biogenesis program, in particular stimulating the activity of the RNA Polymerase I (Pol I) machinery to increase ribosomal RNA (rRNA) transcription. Although MYC function is known to be highly dependent on the cellular signalling context, the pathways interacting with MYC to regulate transcription of ribosomal genes (rDNA) in vivo in response to growth factor status, nutrient availability and cellular stress are only beginning to be understood. To determine factors critical to MYC-dependent stimulation of rDNA transcription in vivo, we performed a transient expression screen for known oncogenic signalling pathways in Drosophila. Strikingly, from the broad range of pathways tested, we found that ribosomal protein S6 Kinase (S6K) activity, downstream of the TOR pathway, was the only factor rate-limiting for the rapid induction of rDNA transcription due to transiently increased MYC. Further, we demonstrated that one of the mechanism(s) by which MYC and S6K cooperate is through coordinate activation of the essential Pol I transcription initiation factor TIF-1A (RRN 3). As Pol I targeted therapy is now in phase 1 clinical trials in patients with haematological malignancies, including those driven by MYC, these data suggest that therapies dually targeting Pol I transcription and S6K activity may be effective in treating MYC-driven tumours.


Subject(s)
DNA, Ribosomal/genetics , Drosophila melanogaster/genetics , Proto-Oncogene Proteins c-myc/physiology , Ribosomal Protein S6 Kinases/physiology , Transcription, Genetic , Animals , Cell Nucleolus/enzymology , Cell Nucleolus/ultrastructure , Compound Eye, Arthropod/enzymology , Compound Eye, Arthropod/ultrastructure , DNA, Ribosomal/metabolism , Drosophila melanogaster/metabolism , Nuclear Proteins/metabolism , Salivary Glands/enzymology , Salivary Glands/ultrastructure , Transcription Factors/metabolism
18.
Nucleic Acids Res ; 43(10): 4975-89, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25916852

ABSTRACT

Nucleoli are not only organelles that produce ribosomal subunits. They are also overarching sensors of different stress conditions and they control specific nucleolar stress pathways leading to stabilization of p53. During DNA replication, ATR and its activator TopBP1 initiate DNA damage response upon DNA damage and replication stress. We found that a basal level of TopBP1 protein associates with ribosomal DNA repeat. When upregulated, TopBP1 concentrates at the ribosomal chromatin and initiates segregation of nucleolar components--the hallmark of nucleolar stress response. TopBP1-induced nucleolar segregation is coupled to shut-down of ribosomal RNA transcription in an ATR-dependent manner. Nucleolar segregation induced by TopBP1 leads to a moderate elevation of p53 protein levels and to localization of activated p53 to nucleolar caps containing TopBP1, UBF and RNA polymerase I. Our findings demonstrate that TopBP1 and ATR are able to inhibit the synthesis of rRNA and to activate nucleolar stress pathway; yet the p53-mediated cell cycle arrest is thwarted in cells expressing high levels of TopBP1. We suggest that inhibition of rRNA transcription by different stress regulators is a general mechanism for cells to initiate nucleolar stress pathway.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleolus/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Ribosomal/biosynthesis , Transcription, Genetic , Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/chemistry , Cell Cycle Checkpoints , Cell Line , Cell Nucleolus/enzymology , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , DNA, Ribosomal/chemistry , DNA-Binding Proteins/chemistry , Humans , Nuclear Proteins/chemistry , Protein Structure, Tertiary , RNA, Ribosomal/genetics , Repetitive Sequences, Nucleic Acid
19.
Nucleic Acids Res ; 42(14): 9005-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25034690

ABSTRACT

DNA topoisomerase II (topo II) changes DNA topology by cleavage/re-ligation cycle(s) and thus contributes to various nuclear DNA transactions. It is largely unknown how the enzyme is controlled in a nuclear context. Several studies have suggested that its C-terminal domain (CTD), which is dispensable for basal relaxation activity, has some regulatory influence. In this work, we examined the impact of nuclear localization on regulation of activity in nuclei. Specifically, human cells were transfected with wild-type and mutant topo IIß tagged with EGFP. Activity attenuation experiments and nuclear localization data reveal that the endogenous activity of topo IIß is correlated with its subnuclear distribution. The enzyme shuttles between an active form in the nucleoplasm and a quiescent form in the nucleolus in a dynamic equilibrium. Mechanistically, the process involves a tethering event with RNA. Isolated RNA inhibits the catalytic activity of topo IIß in vitro through the interaction with a specific 50-residue region of the CTD (termed the CRD). Taken together, these results suggest that both the subnuclear distribution and activity regulation of topo IIß are mediated by the interplay between cellular RNA and the CRD.


Subject(s)
Cell Nucleus/enzymology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , RNA/metabolism , Animals , Biocatalysis , Cell Line , Cell Nucleolus/enzymology , DNA Topoisomerases, Type II/chemistry , DNA, Superhelical/metabolism , DNA-Binding Proteins/chemistry , Humans , Interphase , Mice , Protein Structure, Tertiary , Rats
20.
Acta Histochem ; 116(5): 838-43, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24642039

ABSTRACT

In this study, wheat (Triticum aestivum L.) seeds were used to detect the effect of wortmannin, a specific inhibitor of PI3K, on the nucleolar structure and function. When the germinated seeds were treated with wortmannin, it was shown that the root growth was suppressed and the mitotic index was decreased. The inhibition effects were positively correlated with the concentrations of the drug. The observations of light and transmission electron microscopy revealed that the nucleolar morphology became irregular and their fine structure disappeared. Some granules with a size range of 0.05-0.30 µm diffused from the nucleoli and gradually moved to the nucleoplasm between or around the chromatin. Indirect immunofluorescence staining indicated that B23 shuttled from the nucleoli to the nucleoplasm, or even, to the cytoplasm. RT-PCR technique demonstrated that the expression of C23 was severely down-regulated. Our results suggest, for the first time, that wortmannin treatment can not only damage nucleolar structure, but also inhibit its function, implying that PI3K is involved in nucleolar structure and function.


Subject(s)
Cell Nucleolus/enzymology , Meristem/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Triticum/enzymology , Androstadienes/pharmacology , Microscopy, Electron, Transmission , Phosphoinositide-3 Kinase Inhibitors , Polymerase Chain Reaction , Protein Kinase Inhibitors/pharmacology , Triticum/cytology , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL