Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Biochem Biophys Res Commun ; 636(Pt 2): 113-118, 2022 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-36368153

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Proteínas Pol1 do Complexo de Iniciação de Transcrição , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/patologia , Invasividade Neoplásica/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética
2.
Blood ; 139(24): 3505-3518, 2022 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-35316324

RESUMO

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.


Assuntos
Fator de Transcrição CDX2 , Proteínas Pol1 do Complexo de Iniciação de Transcrição , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Fatores de Transcrição , Adulto , Fator de Transcrição CDX2/genética , Feminino , Genes Homeobox , Humanos , Masculino , Neoplasia Residual/genética , Proteínas de Fusão Oncogênica , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Fatores de Transcrição/genética
3.
PLoS Genet ; 18(2): e1009644, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35139074

RESUMO

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.


Assuntos
Proteínas Pol1 do Complexo de Iniciação de Transcrição , RNA Polimerase I , Animais , Criança , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Humanos , Camundongos , Nucleossomos , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Regiões Promotoras Genéticas , RNA Polimerase I/genética , RNA Ribossômico/genética , Transcrição Gênica
4.
Aging (Albany NY) ; 12(21): 22174-22198, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33146634

RESUMO

Senescence of smooth muscle cells (SMCs) has a crucial role in the pathogenesis of abdominal aortic aneurysm (AAA), a disease of vascular degeneration. Perturbation of cellular ribosomal DNA (rDNA) transcription triggers nucleolar stress response. Previously we demonstrated that induction of nucleolar stress in SMCs elicited cell cycle arrest via the ataxia-telangiectasia mutated (ATM)/ATM- and Rad3-related (ATR)-p53 axis. However, the specific roles of nucleolar stress in vascular degeneration remain unexplored. In the present study, we demonstrated for the first time that in both human and animal AAA tissues, there were non-coordinated changes in the expression of RNA polymerase I machinery components, including a downregulation of transcription initiation factor-IA (TIF-IA). Genetic deletion of TIF-IA in SMCs in mice (smTIF-IA-/-) caused spontaneous aneurysm-like lesions in the aorta. In vitro, induction of nucleolar stress triggered a non-canonical DNA damage response, leading to p53 phosphorylation and a senescence-like phenotype in SMCs. In human AAA tissues, there was increased nucleolar stress in medial cells, accompanied by localized DNA damage response within the nucleolar compartment. Our data suggest that perturbed rDNA transcription and induction of nucleolar stress contribute to the pathogenesis of AAA. Moreover, smTIF-IA-/- mice may be a novel animal model for studying spontaneous AAA-like vascular degenerations.


Assuntos
Nucléolo Celular/patologia , Proliferação de Células , Senescência Celular , Dano ao DNA , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Animais , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Nucléolo Celular/genética , Nucléolo Celular/metabolismo , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fenótipo , Fosforilação , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo
5.
Clin Cancer Res ; 26(17): 4503-4510, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32540851

RESUMO

PURPOSE: DNA mismatch repair (MMR) deficiency is a hallmark of Lynch syndrome, the most common inherited cancer syndrome. MMR-deficient cancer cells accumulate numerous insertion/deletion mutations at microsatellites. Mutations of coding microsatellites (cMS) lead to the generation of immunogenic frameshift peptide (FSP) neoantigens. As the evolution of MMR-deficient cancers is triggered by mutations inactivating defined cMS-containing tumor suppressor genes, distinct FSP neoantigens are shared by most MMR-deficient cancers. To evaluate safety and immunogenicity of an FSP-based vaccine, we performed a clinical phase I/IIa trial (Micoryx). PATIENTS AND METHODS: The trial comprised three cycles of four subcutaneous vaccinations (FSP neoantigens derived from mutant AIM2, HT001, TAF1B genes) mixed with Montanide ISA-51 VG over 6 months. Inclusion criteria were history of MMR-deficient colorectal cancer (UICC stage III or IV) and completion of chemotherapy. Phase I evaluated safety and toxicity as primary endpoint (six patients), phase IIa addressed cellular and humoral immune responses (16 patients). RESULTS: Vaccine-induced humoral and cellular immune responses were observed in all patients vaccinated per protocol. Three patients developed grade 2 local injection site reactions. No vaccination-induced severe adverse events occurred. One heavily pretreated patient with bulky metastases showed stable disease and stable CEA levels over 7 months. CONCLUSIONS: FSP neoantigen vaccination is systemically well tolerated and consistently induces humoral and cellular immune responses, thus representing a promising novel approach for treatment and even prevention of MMR-deficient cancer.


Assuntos
Antígenos de Neoplasias/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Neoplasias Colorretais Hereditárias sem Polipose/terapia , Neoplasias Colorretais/prevenção & controle , Reparo de Erro de Pareamento de DNA , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Neoplasias/efeitos adversos , Antígenos de Neoplasias/genética , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais Hereditárias sem Polipose/complicações , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais Hereditárias sem Polipose/imunologia , Proteínas de Ligação a DNA/genética , Feminino , Mutação da Fase de Leitura , Humanos , Injeções Subcutâneas , Masculino , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Proteínas/genética , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/efeitos adversos , Vacinas de Subunidades Antigênicas/genética
6.
FASEB J ; 34(8): 10818-10834, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32598531

RESUMO

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.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Cromatina/genética , Glucose/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Ribossomos/genética , Transcrição Gênica/genética , Adenosina Trifosfatases/genética , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/genética , RNA Polimerase I/genética , Sirtuínas/genética , Fatores de Transcrição/genética
7.
Eur Rev Med Pharmacol Sci ; 24(9): 4775-4784, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32432789

RESUMO

OBJECTIVE: Previous studies have demonstrated that TNFAIP8 is a cancer-promoting gene. However, the role of TNFAIP8 in esophageal cancer (ECa) has not been reported. The aim of this investigation was to investigate the expression of TNFAIP8 in ECa, and to further explore whether it could regulate cisplatin resistance to this cancer via modulating TAF-Iα expression and promote malignant progression of ECa. PATIENTS AND METHODS: Quantitative Real Time-PCR (qRT-PCR) was performed to examine the expression of TNFAIP8 in 47 tumor tissue specimens and adjacent ones of ECa patients, and the interplay between TNFAIP8 expression and prognosis of patients with ECa was then analyzed. Further, qRT-PCR was applied to verify TNFAIP8 level in ECa cell lines. In addition, the TNFAIP8 knockdown model was constructed in ECa cisplatin-resistant cell lines including EC-109/DDP and OE19/DDP, and then, CCK8 and transwell assays were performed to analyze the impact of TNFAIP8 on the biological function of ECa cells; meanwhile, the Luciferase reporter gene assay and cell reverse experiments were finally conducted to explore its underlying mechanisms. RESULTS: The qRT-PCR results revealed that the TNFAIP8 level in tumor tissue samples of ECa patients was remarkably higher than that in adjacent ones, and the difference was statistically significant. Similarly, the overall survival rate of patients with high expression of TNFAIP8 was lower when compared with patients with low expression of TNFAIP8. EC-109/DDP and OE19/DDP, the ECa cisplatin-resistant cell lines, were successfully constructed; subsequently, it was found that the proliferation, invasiveness, and metastasis ability of ECa cells in TNFAIP8 knockdown group was remarkably decreased compared with those in the sh-NC group. At the same time, the Western blot results illustrated that the expression of TAF-Iαwas remarkably elevated in the TNFAIP8 knockdown group. In addition, the Luciferase reporting assay and cell reverse experiments also demonstrated that there existed a mutual regulation effect between TNFAIP8 and TAF-Iα, which might together affect the malignant progression of ECa. CONCLUSIONS: The expression of TNFAIP8 was found remarkably enhanced in ECa tissues and cell lines, which might be closely relevant to the poor prognosis of patients with ECa. Additionally, it was found that TNFAIP8 may regulate cisplatin resistance and promote malignant progression of ECa by modulating TAF-Iα expression.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas Reguladoras de Apoptose/biossíntese , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Esofágicas/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/biossíntese , Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genética , Humanos , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética
8.
FASEB J ; 34(6): 8283-8295, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32323360

RESUMO

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.


Assuntos
Nucléolo Celular/genética , DNA Ribossômico/genética , Desenvolvimento Embrionário/genética , Ribossomos/genética , Transcrição Gênica/genética , Animais , Blastocisto/fisiologia , Clonagem de Organismos/métodos , Embrião de Mamíferos/fisiologia , Feminino , Camundongos , Técnicas de Transferência Nuclear , Biogênese de Organelas , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Serina-Treonina Quinases TOR/genética
9.
BMC Neurol ; 20(1): 17, 2020 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-31931739

RESUMO

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.


Assuntos
Doenças Neurodegenerativas/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Criança , Variação Genética , Genótipo , Humanos , Masculino , Fenótipo
10.
J Biol Chem ; 294(50): 19155-19166, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31694914

RESUMO

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.


Assuntos
DNA Ribossômico/genética , Regulação para Baixo/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Proteínas Proto-Oncogênicas c-myc/genética , DNA Ribossômico/metabolismo , Perfilação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/metabolismo , Sumoilação , Ativação Transcricional/genética
11.
J Cell Sci ; 132(19)2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31492757

RESUMO

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.


Assuntos
Nucléolo Celular/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , RNA Ribossômico/genética , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , DNA Ribossômico/genética , Humanos , Região Organizadora do Nucléolo/genética , Região Organizadora do Nucléolo/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Transcrição Gênica/genética
12.
J Struct Biol ; 208(2): 191-204, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31479756

RESUMO

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.


Assuntos
Tomografia com Microscopia Eletrônica/métodos , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestrutura , Linhagem Celular , Humanos , Microscopia de Fluorescência , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética
13.
PLoS Genet ; 15(5): e1008157, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31136569

RESUMO

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.


Assuntos
Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , RNA Polimerase I/genética , DNA Ribossômico/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Precursores de RNA/genética , RNA Ribossômico , Ribossomos/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Fatores de Transcrição/genética , Transcrição Gênica
14.
Stem Cell Reports ; 12(5): 1069-1083, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31031188

RESUMO

The Polymerase Associated Factor 1 complex (PAF1c) functions at the interface of epigenetics and gene transcription. The PAF1c is required for MLL fusion-driven acute myeloid leukemia (AML) through direct regulation of pro-leukemic target genes such as Hoxa9 and Meis1. However, the role of the PAF1c in normal hematopoiesis is unknown. Here, we discovered that the PAF1c subunit, CDC73, is required for both fetal and adult hematopoiesis. Loss of Cdc73 in hematopoietic cells is lethal because of extensive bone marrow failure. Cdc73 has an essential cell-autonomous role for adult hematopoietic stem cell function in vivo, and deletion of Cdc73 results in cell-cycle defects in hematopoietic progenitors. Gene expression profiling indicated a differential regulation of Hoxa9/Meis1 gene programs by CDC73 in progenitors compared with AML cells, suggesting disease-specific functions. Thus, the PAF1c subunit, CDC73 is essential for hematopoietic stem cell function but exhibits leukemia-specific regulation of self-renewal gene programs in AML cells.


Assuntos
Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Doença Aguda , Animais , Linhagem Celular Tumoral , Feto/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína Meis1/genética , Proteína Meis1/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo
15.
Cell Death Dis ; 10(3): 196, 2019 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-30814495

RESUMO

ß-dystroglycan (ß-DG) is a key component of multiprotein complexes in the plasma membrane and nuclear envelope. In addition, ß-DG undergoes two successive proteolytic cleavages that result in the liberation of its intracellular domain (ICD) into the cytosol and nucleus. However, stimuli-inducing ICD cleavage and the physiological relevance of this proteolytic fragment are largely unknown. In this study we show for the first time that ß-DG ICD is targeted to the nucleolus where it interacts with the nuclear proteins B23 and UBF (central factor of Pol I-mediated rRNA gene transcription) and binds to rDNA promoter regions. Interestingly DG silencing results in reduced B23 and UBF levels and aberrant nucleolar morphology. Furthermore, ß-DG ICD cleavage is induced by different nucleolar stressors, including oxidative stress, acidosis, and UV irradiation, which implies its participation in the response to nucleolar stress. Consistent with this idea, overexpression of ß-DG elicited mislocalization and decreased levels of UBF and suppression of rRNA expression, which in turn provoked altered ribosome profiling and decreased cell growth. Collectively our data reveal that ß-DG ICD acts as negative regulator of rDNA transcription by impeding the transcriptional activity of UBF, as a part of the protective mechanism activated in response to nucleolar stress.


Assuntos
Nucléolo Celular/metabolismo , Distroglicanas/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , RNA Ribossômico/biossíntese , Animais , Proliferação de Células/genética , Citoplasma/metabolismo , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Distroglicanas/antagonistas & inibidores , Distroglicanas/genética , Camundongos , Mioblastos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Estresse Oxidativo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Domínios Proteicos/genética , RNA Ribossômico/genética , Ribossomos/metabolismo , Transcrição Gênica , Regulação para Cima/genética
16.
PLoS Genet ; 15(2): e1008006, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30802237

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , RNA Polimerase I/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Sequência de Aminoácidos , Proteínas de Ciclo Celular/genética , Sequência Conservada , Humanos , Proteínas Nucleares/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/química , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , RNA Ribossômico/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Deleção de Sequência , Transcrição Gênica
17.
Neuropediatrics ; 50(1): 57-60, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30517966

RESUMO

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.


Assuntos
Epilepsia Resistente a Medicamentos/genética , Mutação/genética , Doenças Neurodegenerativas/genética , Fenótipo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Adolescente , Epilepsia Resistente a Medicamentos/complicações , Epilepsia Resistente a Medicamentos/diagnóstico por imagem , Humanos , Masculino , Doenças Neurodegenerativas/complicações , Doenças Neurodegenerativas/diagnóstico por imagem
18.
RNA Biol ; 15(10): 1286-1294, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30269676

RESUMO

A wide variety of factors are required for the conversion of pre-tRNA molecules into the mature tRNAs that function in translation. To identify factors influencing tRNA biogenesis, we previously performed a screen for strains carrying mutations that induce lethality when combined with a sup61-T47:2C allele, encoding a mutant form of [Formula: see text]. Analyzes of two complementation groups led to the identification of Tan1 as a protein involved in formation of the modified nucleoside N4-acetylcytidine (ac4C) in tRNA and Bud13 as a factor controlling the levels of ac4C by promoting TAN1 pre-mRNA splicing. Here, we describe the remaining complementation groups and show that they include strains with mutations in genes for known tRNA biogenesis factors that modify (DUS2, MOD5 and TRM1), transport (LOS1), or aminoacylate (SES1) [Formula: see text]. Other strains carried mutations in genes for factors involved in rRNA/mRNA synthesis (RPA49, RRN3 and MOT1) or magnesium uptake (ALR1). We show that mutations in not only DUS2, LOS1 and SES1 but also in RPA49, RRN3 and MOT1 cause a reduction in the levels of the altered [Formula: see text]. These results indicate that Rpa49, Rrn3 and Mot1 directly or indirectly influence [Formula: see text] biogenesis.


Assuntos
Adenosina Trifosfatases/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Biossíntese de Proteínas , RNA de Transferência/biossíntese , Proteínas de Saccharomyces cerevisiae/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Alquil e Aril Transferases/genética , Proteínas de Transporte/genética , Mutação , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Oxirredutases/genética , Precursores de RNA/biossíntese , Precursores de RNA/genética , RNA de Transferência/genética , Saccharomyces cerevisiae/genética , tRNA Metiltransferases/genética
19.
Biochem Biophys Res Commun ; 505(1): 325-332, 2018 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-30249398

RESUMO

Spindlin1 (SPIN1), a histone modification reader protein, was enriched in the cell nucleolus and facilitated rRNA expression. However, how SPIN1 localizes to the nucleolus and its functional role in rRNA gene expression remain unresolved. Here, we identified a nucleolar localization signal in the N-terminal region of SPIN1 that is essential for its enrichment and function in the nucleolus. We also discovered that, in addition to its H3K4me3 recognizing activity, the H3R8me2a-recognizing capacity of SPIN1 is also indispensable for stimulating rRNA expression. Chromatin immunoprecipitation results indicated that SPIN1 is required for the association or assembly of selective factor 1 (SL1) complex, probably facilitating the initiation of rDNA transcription through its H3 K4me3-R8me2a reader function.


Assuntos
Proteínas de Ciclo Celular/genética , Nucléolo Celular/genética , Expressão Gênica , Genes de RNAr/genética , Histonas/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Fosfoproteínas/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Imunoprecipitação da Cromatina , Células HEK293 , Células HeLa , Humanos , Metilação , Proteínas Associadas aos Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Ligação Proteica , Interferência de RNA , Transdução de Sinais/genética
20.
Hum Mol Genet ; 27(18): 3154-3164, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29893852

RESUMO

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.


Assuntos
Neoplasias/genética , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , Ribossomos/genética , Proteínas de Transporte Vesicular/genética , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/genética , Carcinogênese/genética , Retículo Endoplasmático/genética , Estresse do Retículo Endoplasmático/genética , Regulação Neoplásica da Expressão Gênica , Mutação em Linhagem Germinativa/genética , Complexo de Golgi/genética , Heterozigoto , Humanos , Mutação com Perda de Função/genética , Neoplasias/patologia , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...