Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Genet ; 18(2): e1009644, 2022 02.
Article in English | MEDLINE | ID: mdl-35139074

ABSTRACT

Transcription of the ~200 mouse and human ribosomal RNA genes (rDNA) by RNA Polymerase I (RPI/PolR1) accounts for 80% of total cellular RNA, around 35% of all nuclear RNA synthesis, and determines the cytoplasmic ribosome complement. It is therefore a major factor controlling cell growth and its misfunction has been implicated in hypertrophic and developmental disorders. Activation of each rDNA repeat requires nucleosome replacement by the architectural multi-HMGbox factor UBTF to create a 15.7 kbp nucleosome free region (NFR). Formation of this NFR is also essential for recruitment of the TBP-TAFI factor SL1 and for preinitiation complex (PIC) formation at the gene and enhancer-associated promoters of the rDNA. However, these promoters show little sequence commonality and neither UBTF nor SL1 display significant DNA sequence binding specificity, making what drives PIC formation a mystery. Here we show that cooperation between SL1 and the longer UBTF1 splice variant generates the specificity required for rDNA promoter recognition in cell. We find that conditional deletion of the TAF1B subunit of SL1 causes a striking depletion of UBTF at both rDNA promoters but not elsewhere across the rDNA. We also find that while both UBTF1 and -2 variants bind throughout the rDNA NFR, only UBTF1 is present with SL1 at the promoters. The data strongly suggest an induced-fit model of RPI promoter recognition in which UBTF1 plays an architectural role. Interestingly, a recurrent UBTF-E210K mutation and the cause of a pediatric neurodegeneration syndrome provides indirect support for this model. E210K knock-in cells show enhanced levels of the UBTF1 splice variant and a concomitant increase in active rDNA copies. In contrast, they also display reduced rDNA transcription and promoter recruitment of SL1. We suggest the underlying cause of the UBTF-E210K syndrome is therefore a reduction in cooperative UBTF1-SL1 promoter recruitment that may be partially compensated by enhanced rDNA activation.


Subject(s)
Pol1 Transcription Initiation Complex Proteins , RNA Polymerase I , Animals , Child , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Humans , Mice , Nucleosomes , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , Promoter Regions, Genetic , RNA Polymerase I/genetics , RNA, Ribosomal/genetics , Transcription, Genetic
2.
Blood ; 139(24): 3505-3518, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35316324

ABSTRACT

Oncogenic alterations underlying B-cell acute lymphoblastic leukemia (B-ALL) in adults remain incompletely elucidated. To uncover novel oncogenic drivers, we performed RNA sequencing and whole-genome analyses in a large cohort of unresolved B-ALL. We identified a novel subtype characterized by a distinct gene expression signature and the unique association of 2 genomic microdeletions. The 17q21.31 microdeletion resulted in a UBTF::ATXN7L3 fusion transcript encoding a chimeric protein. The 13q12.2 deletion resulted in monoallelic ectopic expression of the homeobox transcription factor CDX2, located 138 kb in cis from the deletion. Using 4C-sequencing and CRISPR interference experiments, we elucidated the mechanism of CDX2 cis-deregulation, involving PAN3 enhancer hijacking. CDX2/UBTF ALL (n = 26) harbored a distinct pattern of additional alterations including 1q gain and CXCR4 activating mutations. Within adult patients with Ph- B-ALL enrolled in GRAALL trials, patients with CDX2/UBTF ALL (n = 17/723, 2.4%) were young (median age, 31 years) and dramatically enriched in females (male/female ratio, 0.2, P = .002). They commonly presented with a pro-B phenotype ALL and moderate blast cell infiltration. They had poor response to treatment including a higher risk of failure to first induction course (19% vs 3%, P = .017) and higher post-induction minimal residual disease (MRD) levels (MRD ≥ 10-4, 93% vs 46%, P < .001). This early resistance to treatment translated into a significantly higher cumulative incidence of relapse (75.0% vs 32.4%, P = .004) in univariate and multivariate analyses. In conclusion, we discovered a novel B-ALL entity defined by the unique combination of CDX2 cis-deregulation and UBTF::ATXN7L3 fusion, representing a high-risk disease in young adults.


Subject(s)
CDX2 Transcription Factor , Pol1 Transcription Initiation Complex Proteins , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Transcription Factors , Adult , CDX2 Transcription Factor/genetics , Female , Genes, Homeobox , Humans , Male , Neoplasm, Residual/genetics , Oncogene Proteins, Fusion , Pol1 Transcription Initiation Complex Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Transcription Factors/genetics
3.
Biochem Biophys Res Commun ; 636(Pt 2): 113-118, 2022 12 25.
Article in English | MEDLINE | ID: mdl-36368153

ABSTRACT

Despite tremendous advances in the diagnosis and treatment of NSCLC, the morbidity and mortality of NSCLC still rank high worldwide. Epithelial-mesenchymal transition (EMT) is vital to the invasion, metastasis, and recurrence of NSCLC. Unfortunately, the mechanism behind NSCLC cancer cell EMT remains elusive. Therefore, determining the potential key molecules that induce EMT is important. TATA-binding protein-associated factor-1 (TAF1) is an important component of the preinitiation complex (PIC) that is dysregulated in carcinogenesis. However, the role of TAF1 in NSCLC development is unknown. Therefore, we studied the role of TAF1 in the pathogenesis of NSCLC. First, the expression of TAF1 was determined in human NSCLC tissues and cell lines. TAF1-overexpressing and TAF1 knockdown cell lines were established to evaluate the effect of TAF1 on NSCLC cell proliferation, invasion and migration by colony formation and Transwell assays. The target genes of TAF1 were identified by PCR array and verified by luciferase reporter assay. Our data demonstrated that TAF1 is upregulated in NSCLC. Higher TAF1 expression predicted poor outcomes in NSCLC patients. Mechanistically, TAF1 transcriptionally activated TGFß1, thus promoting NSCLC cell EMT and the development of NSCLC. Targeting TAF1/TGFß1 signalling may be potentially helpful as a therapeutic for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Pol1 Transcription Initiation Complex Proteins , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Neoplasm Invasiveness/genetics , Pol1 Transcription Initiation Complex Proteins/genetics
4.
PLoS Genet ; 15(5): e1008157, 2019 05.
Article in English | MEDLINE | ID: mdl-31136569

ABSTRACT

Most transcriptional activity of exponentially growing cells is carried out by the RNA Polymerase I (Pol I), which produces a ribosomal RNA (rRNA) precursor. In budding yeast, Pol I is a multimeric enzyme with 14 subunits. Among them, Rpa49 forms with Rpa34 a Pol I-specific heterodimer (homologous to PAF53/CAST heterodimer in human Pol I), which might be responsible for the specific functions of the Pol I. Previous studies provided insight in the involvement of Rpa49 in initiation, elongation, docking and releasing of Rrn3, an essential Pol I transcription factor. Here, we took advantage of the spontaneous occurrence of extragenic suppressors of the growth defect of the rpa49 null mutant to better understand the activity of Pol I. Combining genetic approaches, biochemical analysis of rRNA synthesis and investigation of the transcription rate at the individual gene scale, we characterized mutated residues of the Pol I as novel extragenic suppressors of the growth defect caused by the absence of Rpa49. When mapped on the Pol I structure, most of these mutations cluster within the jaw-lobe module, at an interface formed by the lobe in Rpa135 and the jaw made up of regions of Rpa190 and Rpa12. In vivo, the suppressor allele RPA135-F301S restores normal rRNA synthesis and increases Pol I density on rDNA genes when Rpa49 is absent. Growth of the Rpa135-F301S mutant is impaired when combined with exosome mutation rrp6Δ and it massively accumulates pre-rRNA. Moreover, Pol I bearing Rpa135-F301S is a hyper-active RNA polymerase in an in vitro tailed-template assay. We conclude that RNA polymerase I can be engineered to produce more rRNA in vivo and in vitro. We propose that the mutated area undergoes a conformational change that supports the DNA insertion into the cleft of the enzyme resulting in a super-active form of Pol I.


Subject(s)
Pol1 Transcription Initiation Complex Proteins/genetics , RNA Polymerase I/genetics , DNA, Ribosomal/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Precursors/genetics , RNA, Ribosomal , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic
5.
PLoS Genet ; 15(2): e1008006, 2019 02.
Article in English | MEDLINE | ID: mdl-30802237

ABSTRACT

RNA polymerase I (Pol I) synthesizes ribosomal RNA (rRNA) in all eukaryotes, accounting for the major part of transcriptional activity in proliferating cells. Although basal Pol I transcription factors have been characterized in diverse organisms, the molecular basis of the robust rRNA production in vivo remains largely unknown. In S. cerevisiae, the multifunctional Net1 protein was reported to stimulate Pol I transcription. We found that the Pol I-stimulating function can be attributed to the very C-terminal region (CTR) of Net1. The CTR was required for normal cell growth and Pol I recruitment to rRNA genes in vivo and sufficient to promote Pol I transcription in vitro. Similarity with the acidic tail region of mammalian Pol I transcription factor UBF, which could partly functionally substitute for the CTR, suggests conserved roles for CTR-like domains in Pol I transcription from yeast to human.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA Polymerase I/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Amino Acid Sequence , Cell Cycle Proteins/genetics , Conserved Sequence , Humans , Nuclear Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA, Ribosomal/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Deletion , Transcription, Genetic
6.
J Cell Sci ; 132(19)2019 10 09.
Article in English | MEDLINE | ID: mdl-31492757

ABSTRACT

Nucleoli have attracted interest for their role as cellular stress sensors and as potential targets for cancer treatment. The effect of DNA double-strand breaks (DSBs) in nucleoli on rRNA transcription and nucleolar organisation appears to depend on the agent used to introduce DSBs, DSB frequency and the presence (or not) of DSBs outside the nucleoli. To address the controversy, we targeted nucleoli with carbon ions at the ion microbeam SNAKE. Localized ion irradiation with 1-100 carbon ions per point (about 0.3-30 Gy per nucleus) did not lead to overall reduced ribonucleotide incorporation in the targeted nucleolus or other nucleoli of the same cell. However, both 5-ethynyluridine incorporation and Parp1 protein levels were locally decreased at the damaged nucleolar chromatin regions marked by γH2AX, suggesting localized inhibition of rRNA transcription. This locally restricted transcriptional inhibition was not accompanied by nucleolar segregation, a structural reorganisation observed after inhibition of rRNA transcription by treatment with actinomycin D or UV irradiation. The presented data indicate that even multiple complex DSBs do not lead to a pan-nucleolar response if they affect only a subnucleolar region.


Subject(s)
Cell Nucleolus/metabolism , Pol1 Transcription Initiation Complex Proteins/genetics , RNA, Ribosomal/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA, Ribosomal/genetics , Humans , Nucleolus Organizer Region/genetics , Nucleolus Organizer Region/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Transcription, Genetic/genetics
7.
FASEB J ; 34(6): 8283-8295, 2020 06.
Article in English | MEDLINE | ID: mdl-32323360

ABSTRACT

Ribosomal DNA (rDNA) transcription is a limiting step in ribosome biogenesis, crucial for protein synthesis and cell growth-especially at the early stages of embryonic development-and is regulated in a mammalian target of rapamycin (mTOR)-dependent manner. Our previous report demonstrated that treatment with mTOR inhibitors during artificial embryonic activation improved the development of embryos derived from somatic cell nuclear transfer (SCNT). We hypothesize that inhibition of ribosome biogenesis in somatic cells facilitates reactivation of embryonic nucleolar establishment and ribosome biogenesis in SCNT embryos. Herein, we show that mTOR inhibitors suppressed ribosome biogenesis in somatic cells, and more importantly, improved development potential of SCNT embryos (blastocyst rate, 34% vs 24%). SCNT embryos derived from drug-treated somatic cells exhibited higher levels of 47S, 18S, and 5S rRNAs, upstream binding factor (UBF) mRNA, ribosomal protein S6; they also improved the rebuilding of the nucleolar ultrastructure. In addition, treatment of donor cells with the RNA polymerase I (Pol I) inhibitor cx5461 caused similar effects on SCNT embryos. These results indicated that transient inhibition of rDNA transcription in donor cells facilitated the establishment of functional nucleoli and improved preimplantation development of SCNT embryos.


Subject(s)
Cell Nucleolus/genetics , DNA, Ribosomal/genetics , Embryonic Development/genetics , Ribosomes/genetics , Transcription, Genetic/genetics , Animals , Blastocyst/physiology , Cloning, Organism/methods , Embryo, Mammalian/physiology , Female , Mice , Nuclear Transfer Techniques , Organelle Biogenesis , Pol1 Transcription Initiation Complex Proteins/genetics , TOR Serine-Threonine Kinases/genetics
8.
FASEB J ; 34(8): 10818-10834, 2020 08.
Article in English | MEDLINE | ID: mdl-32598531

ABSTRACT

Regulation of ribosomal transcription is under tight control from environmental stimuli, and this control involves changes in the chromatin structure. The underlying mechanism of how chromatin changes in response to nutrient and energy supply in the cell is still unclear. The chromatin-remodeling complex B-WICH is involved in activating the ribosomal transcription, and we show here that knock down of the B-WICH component WSTF results in cells that do not respond to glucose. The promoter is less accessible, and RNA pol I and its transcription factors SL1/TIF-1B and RRN3/TIF-1A, as well as the proto-oncogene c-MYC and the activating deacetylase SIRT7 do not bind upon glucose stimulation. In contrast, the repressive chromatin state that forms after glucose deprivation is reversible, and RNA pol I factors are recruited. WSTF knock down results in an accumulation of the ATPase CHD4, a component of the NuRD chromatin remodeling complex, which is responsible for establishing a repressive poised state at the promoter. The TTF-1, which binds and affect the binding of the chromatin complexes, is important to control the association of activating chromatin component UBF. We suggest that B-WICH is required to allow for a shift to an active chromatin state upon environmental stimulation, by counteracting the repressive state induced by the NuRD complex.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Glucose/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Ribosomes/genetics , Transcription, Genetic/genetics , Adenosine Triphosphatases/genetics , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , RNA Polymerase I/genetics , Sirtuins/genetics , Transcription Factors/genetics
9.
J Biol Chem ; 294(50): 19155-19166, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31694914

ABSTRACT

Ribosome biogenesis is critical for proliferating cells and requires the coordinated activities of three eukaryotic RNA polymerases. We recently showed that the small ubiquitin-like modifier (SUMO) system controls the global level of RNA polymerase II (Pol II)-controlled transcription in mammalian cells by regulating cyclin-dependent kinase 9 activity. Here, we present evidence that the SUMO system also plays a critical role in the control of Pol I transcription. Using an siRNA-based knockdown approach, we found that multiple SUMO E3 ligases of the PIAS (protein inhibitor of activated STAT) family are involved in SUMO-mediated repression of ribosomal DNA (rDNA) gene transcription. We demonstrate that endogenous SUMO represses rDNA transcription primarily by repressing upstream-binding factor and proto-oncogene c-Myc expression and that ectopic overexpression of SUMO-associated enzymes additionally represses rDNA transcription via c-Myc SUMOylation and its subsequent degradation. The results of our study reveal a critical role of SUMOylation in the control of rDNA transcription, uncover the underlying mechanisms involved, and indicate that the SUMO system coordinates Pol I- and Pol II-mediated transcription in mammalian cells.


Subject(s)
DNA, Ribosomal/genetics , Down-Regulation/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , DNA, Ribosomal/metabolism , Gene Expression Profiling , HEK293 Cells , HeLa Cells , Humans , Pol1 Transcription Initiation Complex Proteins/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/metabolism , Sumoylation , Transcriptional Activation/genetics
10.
Hum Mol Genet ; 27(4): 691-705, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29300972

ABSTRACT

UBTF (upstream binding transcription factor) exists as two isoforms; UBTF1 regulates rRNA transcription by RNA polymerase 1, whereas UBTF2 regulates mRNA transcription by RNA polymerase 2. Herein, we describe 4 patients with very similar patterns of neuroregression due to recurrent de novo mutations in UBTF (GRCh37/hg19, NC_000017.10: g.42290219C > T, NM_014233.3: c.628G > A) resulting in the same amino acid change in both UBTF1 and UBTF2 (p.Glu210Lys [p.E210K]). Disease onset in our cohort was at 2.5 to 3 years and characterized by slow progression of global motor, cognitive and behavioral dysfunction. Notable early features included hypotonia with a floppy gait, high-pitched dysarthria and hyperactivity. Later features included aphasia, dystonia, and spasticity. Speech and ambulatory ability were lost by the early teens. Magnetic resonance imaging showed progressive generalized cerebral atrophy (supratentorial > infratentorial) with involvement of both gray and white matter. Patient fibroblasts showed normal levels of UBTF transcripts, increased expression of pre-rRNA and 18S rRNA, nucleolar abnormalities, markedly increased numbers of DNA breaks, defective cell-cycle progression, and apoptosis. Expression of mutant human UBTF1 in Drosophila neurons was lethal. Although no loss-of-function variants are reported in the Exome Aggregation Consortium (ExAC) database and Ubtf-/- is early embryonic lethal in mice, Ubtf+/- mice displayed only mild motor and behavioral dysfunction in adulthood. Our data underscore the importance of including UBTF E210K in the differential diagnosis of neuroregression and suggest that mainly gain-of-function mechanisms contribute to the pathogenesis of the UBTF E210K neuroregression syndrome.


Subject(s)
Mutation, Missense/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , Child, Preschool , Dysarthria/genetics , Female , Gait Ataxia/genetics , Humans , Magnetic Resonance Imaging , Male , Muscle Hypotonia/genetics , Pedigree , RNA, Ribosomal, 18S/genetics
11.
Hum Mol Genet ; 27(18): 3154-3164, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29893852

ABSTRACT

SEC23B is a component of coat protein complex II (COPII) vesicles that transport secretory proteins from the endoplasmic reticulum (ER) to the Golgi apparatus. Loss-of-function SEC23B mutations cause a rare form of anemia, resulting from decreased SEC23B levels. We recently identified germline heterozygous SEC23B variants as potentially cancer-predisposing. Mutant SEC23B associated with ER stress-mediated tumorigenesis, without decreased SEC23B expression. However, our understanding of the processes behind these observations remain limited. Here, we show mutant SEC23B exists within nucleoli, in addition to classical distribution at the ER/Golgi. This occurs independent of other COPII proteins and does not compromise secretory function. Mutant cells have increased ribosomal protein and translation-related gene expression, and enhanced translational capacity, in the presence of ER stress. We show that mutant SEC23B binds to UBF transcription factor, with increased UBF transcription factor binding at the ribosomal DNA promoter. Our data indicate SEC23B has potential non-canonical COPII-independent function, particularly within the ribosome biogenesis pathway, and that may contribute to the pathogenesis of cancer-predisposition.


Subject(s)
Neoplasms/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , Ribosomes/genetics , Vesicular Transport Proteins/genetics , COP-Coated Vesicles/genetics , Carcinogenesis/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic , Germ-Line Mutation/genetics , Golgi Apparatus/genetics , Heterozygote , Humans , Loss of Function Mutation/genetics , Neoplasms/pathology , Protein Binding
12.
Am J Hum Genet ; 101(2): 267-273, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28777933

ABSTRACT

Ribosomal RNA (rRNA) is transcribed from rDNA by RNA polymerase I (Pol I) to produce the 45S precursor of the 28S, 5.8S, and 18S rRNA components of the ribosome. Two transcription factors have been defined for Pol I in mammals, the selectivity factor SL1, and the upstream binding transcription factor (UBF), which interacts with the upstream control element to facilitate the assembly of the transcription initiation complex including SL1 and Pol I. In seven unrelated affected individuals, all suffering from developmental regression starting at 2.5-7 years, we identified a heterozygous variant, c.628G>A in UBTF, encoding p.Glu210Lys in UBF, which occurred de novo in all cases. While the levels of UBF, Ser388 phosphorylated UBF, and other Pol I-related components (POLR1E, TAF1A, and TAF1C) remained unchanged in cells of an affected individual, the variant conferred gain of function to UBF, manifesting by markedly increased UBF binding to the rDNA promoter and to the 5'- external transcribed spacer. This was associated with significantly increased 18S expression, and enlarged nucleoli which were reduced in number per cell. The data link neurodegeneration in childhood with altered rDNA chromatin status and rRNA metabolism.


Subject(s)
Brain Diseases/genetics , Cell Nucleolus/pathology , Neurodegenerative Diseases/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , RNA, Ribosomal, 18S/biosynthesis , Adolescent , Adult , Atrophy/genetics , Brain/pathology , Brain Diseases/pathology , Child , Chromatin/metabolism , DNA-Binding Proteins/genetics , Female , Humans , Male , Neurodegenerative Diseases/pathology , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Young Adult
13.
Genome Res ; 27(9): 1525-1535, 2017 09.
Article in English | MEDLINE | ID: mdl-28684554

ABSTRACT

Translational efficiency is subject to extensive regulation. However, the factors influencing such regulation are poorly understood. In Caenorhabditis elegans, 62% of genes are trans-spliced to a specific spliced leader (SL1), which replaces part of the native 5' untranslated region (5' UTR). Given the pivotal role the 5' UTR plays in the regulation of translational efficiency, we hypothesized that SL1 trans-splicing functions to regulate translational efficiency. With genome-wide analysis on Ribo-seq data, polysome profiling experiments, and CRISPR-Cas9-based genetic manipulation of trans-splicing sites, we found four lines of evidence in support of this hypothesis. First, SL1 trans-spliced genes have higher translational efficiencies than non-trans-spliced genes. Second, SL1 trans-spliced genes have higher translational efficiencies than non-trans-spliced orthologous genes in other nematode species. Third, an SL1 trans-spliced isoform has higher translational efficiency than the non-trans-spliced isoform of the same gene. Fourth, deletion of trans-splicing sites of endogenous genes leads to reduced translational efficiency. Importantly, we demonstrated that SL1 trans-splicing plays a key role in enhancing translational efficiencies of essential genes. We further discovered that SL1 trans-splicing likely enhances translational efficiency by shortening the native 5' UTRs, hence reducing the presence of upstream start codons (uAUG) and weakening mRNA secondary structures. Taken together, our study elucidates the global function of trans-splicing in enhancing translational efficiency in nematodes, paving the way for further understanding the genomic mechanisms of translational regulation.


Subject(s)
Pol1 Transcription Initiation Complex Proteins/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Trans-Splicing/genetics , 5' Untranslated Regions/genetics , Animals , CRISPR-Cas Systems/genetics , Caenorhabditis elegans/genetics , Codon, Initiator/genetics , Gene Editing , Genome/genetics , RNA Splicing/genetics , RNA, Messenger/biosynthesis
14.
BMC Neurol ; 20(1): 17, 2020 Jan 13.
Article in English | MEDLINE | ID: mdl-31931739

ABSTRACT

BACKGROUND: A new monogenic neurodegenerative disease affecting ribosomal metabolism has recently been identified in association with a monoallelic UBTF putative gain of function variant (NM_001076683.1:c.628G>A, hg19). Phenotype is consistent among these probands with progressive motor, cognitive, and behavioural regression in early to middle childhood. CASE PRESENTATION: We report on a child with this monoallelic UBTF variant who presented with progressive disease including regression, episodes of subacute deterioration during febrile illnesses and a remarkable EEG pattern with a transient pattern of semi-periodic slow waves. CONCLUSIONS: This case further supports the phenotype-genotype correlation of neurodegeneration associated with UBTF c.628G>A. Moreover, it brings new insights into the clinical features and EEG that could possibly serve as diagnostic markers of this otherwise nonspecific phenotype.


Subject(s)
Neurodegenerative Diseases/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , Child , Genetic Variation , Genotype , Humans , Male , Phenotype
15.
PLoS Genet ; 13(7): e1006899, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28715449

ABSTRACT

Transcription of the several hundred of mouse and human Ribosomal RNA (rRNA) genes accounts for the majority of RNA synthesis in the cell nucleus and is the determinant of cytoplasmic ribosome abundance, a key factor in regulating gene expression. The rRNA genes, referred to globally as the rDNA, are clustered as direct repeats at the Nucleolar Organiser Regions, NORs, of several chromosomes, and in many cells the active repeats are transcribed at near saturation levels. The rDNA is also a hotspot of recombination and chromosome breakage, and hence understanding its control has broad importance. Despite the need for a high level of rDNA transcription, typically only a fraction of the rDNA is transcriptionally active, and some NORs are permanently silenced by CpG methylation. Various chromatin-remodelling complexes have been implicated in counteracting silencing to maintain rDNA activity. However, the chromatin structure of the active rDNA fraction is still far from clear. Here we have combined a high-resolution ChIP-Seq protocol with conditional inactivation of key basal factors to better understand what determines active rDNA chromatin. The data resolve questions concerning the interdependence of the basal transcription factors, show that preinitiation complex formation is driven by the architectural factor UBF (UBTF) independently of transcription, and that RPI termination and release corresponds with the site of TTF1 binding. They further reveal the existence of an asymmetric Enhancer Boundary Complex formed by CTCF and Cohesin and flanked upstream by phased nucleosomes and downstream by an arrested RNA Polymerase I complex. We find that the Enhancer Boundary Complex is the only site of active histone modification in the 45kbp rDNA repeat. Strikingly, it not only delimits each functional rRNA gene, but also is stably maintained after gene inactivation and the re-establishment of surrounding repressive chromatin. Our data define a poised state of rDNA chromatin and place the Enhancer Boundary Complex as the likely entry point for chromatin remodelling complexes.


Subject(s)
Genes, rRNA , Pol1 Transcription Initiation Complex Proteins/genetics , RNA Polymerase I/metabolism , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Female , Gene Deletion , Gene Silencing , Genetic Loci , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleolus Organizer Region/genetics , Nucleolus Organizer Region/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , Pregnancy , RNA Polymerase I/genetics , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
16.
J Struct Biol ; 208(2): 191-204, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31479756

ABSTRACT

Upstream binding transcription factor (UBTF) is a co-regulator of RNA polymerase I by constituting an initiation complex on rRNA genes. UBTF plays a role in rDNA bending and its maintenance in "open" state. It exists as two splicing variants, UBTF1 and UBTF2, which cannot be discerned with antibodies raised against UBTF. We investigated the ultrastructural localization of each variant in cells synthesizing GFP-tagged UBTF1 or UBTF2 by using anti-GFP antibodies and pre-embedding nanogold strategy. Detailed 3D distribution of UBTF1 and 2 was also studied by electron tomography. In control cells, the two isoforms are very abundant within fibrillar centers, but their repartition strongly differs. Electron tomography shows that UBTF1 is disposed as fibrils that are folded in coils whereas UBTF2 is localized homogenously, preferentially at their cortical area. As UBTF is a useful marker to trace rDNA genes, we used these data to improve our previous model of 3D organization of active transcribing rDNA gene within fibrillar centers. Finally, when rRNA synthesis is inhibited during actinomycin D treatment or entry in mitosis, UBTF1 and UBTF2 show a similar distribution along extended 3D loop-like structures. Altogether these data suggest new roles for UBTF1 and UBTF2 isoforms in the organization of active and inactive rDNA genes.


Subject(s)
Electron Microscope Tomography/methods , Pol1 Transcription Initiation Complex Proteins/metabolism , Protein Isoforms/metabolism , Protein Isoforms/ultrastructure , Cell Line , Humans , Microscopy, Fluorescence , Pol1 Transcription Initiation Complex Proteins/genetics
17.
J Biol Chem ; 293(7): 2358-2369, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29301938

ABSTRACT

The nucleoli accumulate rRNA genes and are the sites of rRNA synthesis and rRNA assembly into ribosomes. During mitosis, nucleoli dissociate, but nucleolar remnants remain on the rRNA gene loci, forming distinct nucleolar organizer regions (NORs). Little is known about the composition and structure of NORs, but upstream binding factor (UBF) has been established as its master organizer. In this study, we sought to establish new proteins in NORs. Using UBF-Sepharose to isolate UBF-binding proteins, we identified histone H1.2 as a candidate partner but were puzzled by this observation, given that UBF is known to be located predominantly in nucleoli, whereas H1.2 distributed broadly among the chromatins in interphase nuclei. We then examined cells undergoing mitosis and saw that both H1.2 and UBF were recruited into NORs in this state, reconciling the results of our UBF pulldowns. Inhibiting rRNA synthesis in interphase nuclei also induced NOR-like structures containing both UBF and H1.2. When chromosomes were isolated and spread on coverslips, NORs appeared separated from the chromosomes containing both UBF and H1.2. After chromosomes were fragmented by homogenization, intact NORs remained visible. Results collectively suggest that NORs are independent structures and that the linker histone H1.2 is a novel component of this structure.


Subject(s)
Histones/metabolism , Nucleolus Organizer Region/metabolism , Chromatin/genetics , Chromatin/metabolism , Histones/genetics , Humans , Mitosis , Nucleolus Organizer Region/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Transcription, Genetic
18.
Hum Mol Genet ; 26(15): 2874-2881, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28472305

ABSTRACT

Non-ischemic dilated cardiomyopathy (DCM) has been recognized as a heritable disorder for over 25 years, yet clinical genetic testing is non-diagnostic in >50% of patients, underscoring the ongoing need for DCM gene discovery. Here, whole exome sequencing uncovered a novel molecular basis for idiopathic end-stage heart failure in two sisters who underwent cardiac transplantation at three years of age. Compound heterozygous recessive mutations in TAF1A, encoding an RNA polymerase I complex protein, were associated with marked fibrosis of explanted hearts and gene-specific nucleolar segregation defects in cardiomyocytes, indicative of impaired ribosomal RNA synthesis. Knockout of the homologous gene in zebrafish recapitulated a heart failure phenotype with pericardial edema, decreased ventricular systolic function, and embryonic mortality. These findings expand the clinical spectrum of ribosomopathies to include pediatric DCM.


Subject(s)
Cardiomyopathy, Dilated/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , Animals , Child , Child, Preschool , Exome , Female , Fibrosis/genetics , Genetic Testing , Heart Failure , Heterozygote , Humans , Male , Mutation , Mutation, Missense/genetics , Myocytes, Cardiac/metabolism , Pedigree , RNA, Ribosomal/biosynthesis , RNA, Ribosomal/genetics , Siblings , Exome Sequencing , Zebrafish/genetics
19.
Blood ; 129(21): 2882-2895, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28283481

ABSTRACT

Despite the development of novel drugs, the prospects for many patients with acute myeloid leukemia (AML) remain dismal. This study reveals that the selective inhibitor of RNA polymerase I (Pol I) transcription, CX-5461, effectively treats aggressive AML, including mixed-lineage leukemia-driven AML, and outperforms standard chemotherapies. In addition to the previously characterized mechanism of action of CX-5461 (ie, the induction of p53-dependent apoptotic cell death), the inhibition of Pol I transcription also demonstrates potent efficacy in p53null AML in vivo. This significant survival advantage in both p53WT and p53null leukemic mice treated with CX-5461 is associated with activation of the checkpoint kinases 1/2, an aberrant G2/M cell-cycle progression and induction of myeloid differentiation of the leukemic blasts. The ability to target the leukemic-initiating cell population is thought to be essential for lasting therapeutic benefit. Most strikingly, the acute inhibition of Pol I transcription reduces both the leukemic granulocyte-macrophage progenitor and leukemia-initiating cell (LIC) populations, and suppresses their clonogenic capacity. This suggests that dysregulated Pol I transcription is essential for the maintenance of their leukemia-initiating potential. Together, these findings demonstrate the therapeutic utility of this new class of inhibitors to treat highly aggressive AML by targeting LICs.


Subject(s)
Benzothiazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Naphthyridines/pharmacology , Neoplastic Stem Cells/enzymology , Pol1 Transcription Initiation Complex Proteins/antagonists & inhibitors , Transcription, Genetic/drug effects , Animals , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , G2 Phase/drug effects , G2 Phase/genetics , Humans , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, Mutant Strains , Neoplastic Stem Cells/pathology , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Neuropediatrics ; 50(1): 57-60, 2019 02.
Article in English | MEDLINE | ID: mdl-30517966

ABSTRACT

INTRODUCTION: Neurodegenerative diseases of childhood present with progressive decline in cognitive, social, and motor function and are frequently associated with seizures in different stages of the disease. Here we report a patient with severe progressive neurodegeneration with drug-resistant epilepsy of unknown etiology from the age of 2 years. METHODS AND RESULTS: Using whole exome sequencing, we found heterozygous missense de novo variant c.628G > A (p.Glu210Lys) in the UBTF gene. This variant was recently described as de novo in 11 patients with similar neurodegeneration characterized by developmental decline initially confined to motor development followed by language regression, appearance of an extrapyramidal movement disorder, and leading to severe intellectual disability. In 3 of the 11 patients described so far, seizures were also present. CONCLUSIONS: Our report expands the complex phenotype of neurodegeneration associated with the c.628G > A variant in the UBTF gene and helps to clarify the relation between this one single recurrent pathogenic variant described in this gene to date and its phenotype. The UBTF gene should be considered a novel candidate gene in neurodegeneration with or without epilepsy.


Subject(s)
Drug Resistant Epilepsy/genetics , Mutation/genetics , Neurodegenerative Diseases/genetics , Phenotype , Pol1 Transcription Initiation Complex Proteins/genetics , Adolescent , Drug Resistant Epilepsy/complications , Drug Resistant Epilepsy/diagnostic imaging , Humans , Male , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL