Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Hum Genet ; 143(3): 437-453, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38520561

RESUMEN

General transcription factor IIIC subunit 5 (GTF3C5) encodes transcription factor IIIC63 (TFIIIC63). It binds to DNA to recruit another transcription factor, TFIIIB, and RNA polymerase III (Pol III) to mediate the transcription of small noncoding RNAs, such as tRNAs. Here, we report four individuals from three families presenting with a multisystem developmental disorder phenotype with biallelic variants in GTF3C5. The overlapping features include growth retardation, developmental delay, intellectual disability, dental anomalies, cerebellar malformations, delayed bone age, skeletal anomalies, and facial dysmorphism. Using lymphoblastoid cell lines (LCLs) from two affected individuals, we observed a reduction in TFIIIC63 protein levels compared to control LCLs. Genome binding of TFIIIC63 protein is also reduced in LCL from one of the affected individuals. Additionally, approximately 40% of Pol III binding regions exhibited reduction in the level of Pol III occupancy in the mutant genome relative to the control, while approximately 54% of target regions showed comparable levels of Pol III occupancy between the two, indicating partial impairment of Pol III occupancy in the mutant genome. Yeasts with subject-specific variants showed temperature sensitivity and impaired growth, supporting the notion that the identified variants have deleterious effects. gtf3c5 mutant zebrafish showed developmental defects, including a smaller body, head, and eyes. Taken together, our data show that GTF3C5 plays an important role in embryonic development, and that biallelic variants in this gene cause a multisystem developmental disorder. Our study adds GTF3C5-related disorder to the growing list of genetic disorders associated with Pol III transcription machinery.


Asunto(s)
Discapacidades del Desarrollo , ARN Polimerasa III , Factores de Transcripción TFIII , Animales , Niño , Preescolar , Femenino , Humanos , Masculino , Alelos , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Discapacidad Intelectual/genética , Mutación , Linaje , Fenotipo , ARN Polimerasa III/genética , ARN Polimerasa III/metabolismo , Factores de Transcripción TFII/genética , Factores de Transcripción TFII/metabolismo , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/metabolismo , Transcripción Genética , Pez Cebra/genética
2.
Glia ; 72(6): 1117-1135, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38450767

RESUMEN

Williams syndrome (WS) is a genetic neurodevelopmental disorder caused by a heterozygous microdeletion, characterized by hypersociability and unique neurocognitive abnormalities. Of the deleted genes, GTF2I has been linked to hypersociability in WS. We have recently shown that Gtf2i deletion from forebrain excitatory neurons, referred to as Gtf2i conditional knockout (cKO) mice leads to multi-faceted myelination deficits associated with the social behaviors affected in WS. These deficits were potentially mediated also by microglia, as they present a close relationship with oligodendrocytes. To study the impact of altered myelination, we characterized these mice in terms of microglia over the course of development. In postnatal day 30 (P30) Gtf2i cKO mice, cortical microglia displayed a more ramified state, as compared with wild type (controls). However, postnatal day 4 (P4) microglia exhibited high proliferation rates and an elevated activation state, demonstrating altered properties related to activation and inflammation in Gtf2i cKO mice compared with control. Intriguingly, P4 Gtf2i cKO-derived microglial cells exhibited significantly elevated myelin phagocytosis in vitro compared to control mice. Lastly, systemic injection of clemastine to P4 Gtf2i cKO and control mice until P30, led to a significant interaction between genotypes and treatments on the expression levels of the phagocytic marker CD68, and a significant reduction of the macrophage/microglial marker Iba1 transcript levels in the cortex of the Gtf2i cKO treated mice. Our data thus implicate microglia as important players in WS, and that early postnatal manipulation of microglia might be beneficial in treating inflammatory and myelin-related pathologies.


Asunto(s)
Factores de Transcripción TFIII , Factores de Transcripción TFII , Síndrome de Williams , Ratones , Animales , Microglía , Síndrome de Williams/genética , Neuronas/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción TFIII/metabolismo , Factores de Transcripción TFII/genética , Factores de Transcripción TFII/metabolismo
3.
Cell Rep ; 43(3): 113867, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38416640

RESUMEN

Individuals with Williams syndrome (WS), a neurodevelopmental disorder caused by hemizygous loss of 26-28 genes at 7q11.23, characteristically portray a hypersocial phenotype. Copy-number variations and mutations in one of these genes, GTF2I, are associated with altered sociality and are proposed to underlie hypersociality in WS. However, the contribution of GTF2I to human neurodevelopment remains poorly understood. Here, human cellular models of neurodevelopment, including neural progenitors, neurons, and three-dimensional cortical organoids, are differentiated from CRISPR-Cas9-edited GTF2I-knockout (GTF2I-KO) pluripotent stem cells to investigate the role of GTF2I in human neurodevelopment. GTF2I-KO progenitors exhibit increased proliferation and cell-cycle alterations. Cortical organoids and neurons demonstrate increased cell death and synaptic dysregulation, including synaptic structural dysfunction and decreased electrophysiological activity on a multielectrode array. Our findings suggest that changes in synaptic circuit integrity may be a prominent mediator of the link between alterations in GTF2I and variation in the phenotypic expression of human sociality.


Asunto(s)
Factores de Transcripción TFIII , Factores de Transcripción TFII , Síndrome de Williams , Humanos , Síndrome de Williams/genética , Síndrome de Williams/metabolismo , Neuronas/metabolismo , Conducta Social , Fenotipo , Factores de Transcripción TFIII/metabolismo , Factores de Transcripción TFII/genética , Factores de Transcripción TFII/metabolismo
4.
Commun Biol ; 6(1): 1269, 2023 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-38097729

RESUMEN

Gtf2i encodes the general transcription factor II-I (TFII-I), with peak expression during pre-natal and early post-natal brain development stages. Because these stages are critical for proper brain development, we studied at the single-cell level the consequences of Gtf2i's deletion from excitatory neurons, specifically on mitochondria. Here we show that Gtf2i's deletion resulted in abnormal morphology, disrupted mRNA related to mitochondrial fission and fusion, and altered autophagy/mitophagy protein expression. These changes align with elevated reactive oxygen species levels, illuminating Gtf2i's importance in neurons mitochondrial function. Similar mitochondrial issues were demonstrated by Gtf2i heterozygous model, mirroring the human condition in Williams syndrome (WS), and by hemizygous neuronal Gtf2i deletion model, indicating Gtf2i's dosage-sensitive role in mitochondrial regulation. Clinically relevant, we observed altered transcript levels related to mitochondria, hypoxia, and autophagy in frontal cortex tissue from WS individuals. Our study reveals mitochondrial and autophagy-related deficits shedding light on WS and other Gtf2i-related disorders.


Asunto(s)
Factores de Transcripción TFIII , Síndrome de Williams , Humanos , Autofagia/genética , Heterocigoto , Neuronas/metabolismo , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/metabolismo , Síndrome de Williams/genética , Síndrome de Williams/metabolismo
5.
Front Med ; 17(6): 1186-1203, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37707678

RESUMEN

Through bioinformatics predictions, we identified that GTF2I and FAT1 were downregulated in thyroid carcinoma (TC). Further, Pearson's correlation coefficient revealed a positive correlation between GTF2I expression and FAT1 expression. Therefore, we selected them for this present study, where the effects of bone marrow mesenchymal stem cell-derived EVs (BMSDs-EVs) enriched with GTF2I were evaluated on the epithelial-to-mesenchymal transition (EMT) and stemness maintenance in TC. The under-expression of GTF2I and FAT1 was validated in TC cell lines. Ectopically expressed GTF2I and FAT1 were found to augment malignant phenotypes of TC cells, EMT, and stemness maintenance. Mechanistic studies revealed that GTF2I bound to the promoter region of FAT1 and consequently upregulated its expression. MSC-EVs could shuttle GTF2I into TPC-1 cells, where GTF2I inhibited TC malignant phenotypes, EMT, and stemness maintenance by increasing the expression of FAT1 and facilitating the FAT1-mediated CDK4/FOXM1 downregulation. In vivo experiments confirmed that silencing of GTF2I accelerated tumor growth in nude mice. Taken together, our work suggests that GTF2I transferred by MSC-EVs confer antioncogenic effects through the FAT1/CDK4/FOXM1 axis and may be used as a promising biomarker for TC treatment.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Neoplasias de la Tiroides , Factores de Transcripción TFIII , Ratones , Animales , Línea Celular Tumoral , Proliferación Celular , Ratones Desnudos , Transición Epitelial-Mesenquimal , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Vesículas Extracelulares/patología , Factores de Transcripción TFIII/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
6.
Int J Mol Sci ; 24(4)2023 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-36835038

RESUMEN

Regulation of histone acetylation dictates patterns of gene expression and hence cell identity. Due to their clinical relevance in cancer biology, understanding how human embryonic stem cells (hESCs) regulate their genomic patterns of histone acetylation is critical, but it remains largely to be investigated. Here, we provide evidence that acetylation of histone H3 lysine-18 (H3K18ac) and lysine-27 (H3K27ac) is only partially established by p300 in stem cells, while it represents the main histone acetyltransferase (HAT) for these marks in somatic cells. Our analysis reveals that whereas p300 marginally associated with H3K18ac and H3K27ac in hESCs, it largely overlapped with these histone marks upon differentiation. Interestingly, we show that H3K18ac is found at "stemness" genes enriched in RNA polymerase III transcription factor C (TFIIIC) in hESCs, whilst lacking p300. Moreover, TFIIIC was also found in the vicinity of genes involved in neuronal biology, although devoid of H3K18ac. Our data suggest a more complex pattern of HATs responsible for histone acetylations in hESCs than previously considered, suggesting a putative role for H3K18ac and TFIIIC in regulating "stemness" genes as well as genes associated with neuronal differentiation of hESCs. The results break ground for possible new paradigms for genome acetylation in hESCs that could lead to new avenues for therapeutic intervention in cancer and developmental diseases.


Asunto(s)
Epigénesis Genética , Histona Acetiltransferasas , Factores de Transcripción TFIII , Humanos , Acetilación , Células Madre Embrionarias , Epigénesis Genética/fisiología , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Factores de Transcripción TFIII/metabolismo
7.
Front Immunol ; 14: 1067459, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36756127

RESUMEN

The ubiquitously expressed transcription factor TFII-I is a multifunctional protein with pleiotropic roles in gene regulation. TFII-I associated polymorphisms are implicated in Sjögren's syndrome and Lupus in humans and, germline deletion of the Gtf2i gene in mice leads to embryonic lethality. Here we report a unique role for TFII-I in homeostasis of innate properties of B lymphocytes. Loss of Gtf2i in murine B lineage cells leads to an alteration in transcriptome, chromatin landscape and associated transcription factor binding sites, which exhibits myeloid-like features and coincides with enhanced sensitivity to LPS induced gene expression. TFII-I deficient B cells also show increased switching to IgG3, a phenotype associated with inflammation. These results demonstrate a role for TFII-I in maintaining immune homeostasis and provide clues for GTF2I polymorphisms associated with B cell dominated autoimmune diseases in humans.


Asunto(s)
Síndrome de Sjögren , Factores de Transcripción TFIII , Factores de Transcripción TFII , Humanos , Ratones , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cromatina , Unión Proteica , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/metabolismo , Factores de Transcripción TFII/genética , Factores de Transcripción TFII/metabolismo
8.
Genes (Basel) ; 13(10)2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36292679

RESUMEN

Autism spectrum disorder (ASD) is an early onset, developmental disorder whose genetic cause is heterogeneous and complex. In total, 70% of ASD cases are due to an unknown etiology. Among the monogenic causes of ASD, fragile X syndrome (FXS) accounts for 2-4% of ASD cases, and 60% of individuals with FXS present with ASD. Epigenetic changes, specifically DNA methylation, which modulates gene expression levels, play a significant role in the pathogenesis of both disorders. Thus, in this study, using the Human Methylation EPIC Bead Chip, we examined the global DNA methylation profiles of biological samples derived from 57 age-matched male participants (2-6 years old), including 23 subjects with ASD, 23 subjects with FXS with ASD (FXSA) and 11 typical developing (TD) children. After controlling for technical variation and white blood cell composition, using the conservatory threshold of the false discovery rate (FDR ≤ 0.05), in the three comparison groups, TD vs. AD, TD vs. FXSA and ASD vs. FXSA, we identified 156, 79 and 3100 differentially methylated sites (DMS), and 14, 13 and 263 differential methylation regions (DMRs). Interestingly, several genes differentially methylated among the three groups were among those listed in the SFARI Gene database, including the PAK2, GTF2I and FOXP1 genes important for brain development. Further, enrichment analyses identified pathways involved in several functions, including synaptic plasticity. Our preliminary study identified a significant role of altered DNA methylation in the pathology of ASD and FXS, suggesting that the characterization of a DNA methylation signature may help to unravel the pathogenicity of FXS and ASD and may help the development of an improved diagnostic classification of children with ASD and FXSA. In addition, it may pave the way for developing therapeutic interventions that could reverse the altered methylome profile in children with neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista , Síndrome del Cromosoma X Frágil , Factores de Transcripción TFIII , Niño , Humanos , Masculino , Preescolar , Síndrome del Cromosoma X Frágil/genética , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Metilación de ADN/genética , Epigénesis Genética/genética , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/metabolismo , Proteínas Represoras/genética , Factores de Transcripción Forkhead/genética
9.
BMC Genomics ; 23(1): 656, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36114454

RESUMEN

BACKGROUND: General transcription factor IIi (GTF2I) mutations are very common in thymic epithelial tumors (TETs) and are related to a more favorable prognosis in TET patients. However, limited research has been conducted on the role of GTF2I in the tumor immune microenvironment (TIME). Further, long non-coding RNAs (lncRNAs) have been associated with the survival of patients with TETs. Therefore, this study aimed to explore the relationship between GTF2I mutations and TIME and build a new potential signature for predicting tumor recurrence in the TETs. Research data was downloaded from The Cancer Genome Atlas database and the CIBERSORT algorithm was used to evaluate TIME differences between GTF2I mutant and wild-type TETs. Relevant differentially expressed lncRNAs based on differentially expressed immune-related genes were identified to establish lncRNA pairs. We constructed a signature using univariate and multivariate Cox regression analyses. RESULTS: GTF2I is the most commonly mutated gene in TETs, and is associated with an increased number of early-stage pathological types, as well as no history of myasthenia gravis or radiotherapy treatment. In the GTF2I wild-type group, immune score and immune cell infiltrations with M2 macrophages, activated mast cells, neutrophils, plasma, T helper follicular cells, and activated memory CD4 T cells were higher than the GTF2I mutant group. A risk model was built using five lncRNA pairs, and the 1-, 3-, and 5-year area under the curves were 0.782, 0.873, and 0.895, respectively. A higher risk score was related to more advanced histologic type. CONCLUSION: We can define the GTF2I mutant-type TET as an immune stable type and the GTF2I wild-type as an immune stressed type. A signature based on lncRNA pairs was also constructed to effectively predict tumor recurrence.


Asunto(s)
Neoplasias Glandulares y Epiteliales , ARN Largo no Codificante , Factores Generales de Transcripción , Factores de Transcripción TFIII , Factores de Transcripción TFII , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Recurrencia Local de Neoplasia/genética , Neoplasias Glandulares y Epiteliales/genética , Pronóstico , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias del Timo , Factores Generales de Transcripción/genética , Factores Generales de Transcripción/metabolismo , Factores de Transcripción TFII/genética , Factores de Transcripción TFII/metabolismo , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/metabolismo , Microambiente Tumoral
10.
J Biol Chem ; 296: 100456, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33636181

RESUMEN

The disease-initiating molecular events for age-related macular degeneration (AMD), a multifactorial retinal disease affecting many millions of elderly individuals worldwide, are still unknown. Of the over 30 risk and protective loci so far associated with AMD through whole genome-wide association studies (GWAS), the Age-Related Maculopathy Susceptibility 2 (ARMS2) gene locus represents one of the most highly associated risk regions for AMD. A unique insertion/deletion (in/del) sequence located immediately upstream of the High Temperature Requirement A1 (HTRA1) gene in this region confers high risk for AMD. Using electrophoretic mobility shift assay (EMSA), we identified that two Gtf2i-ß/δ transcription factor isoforms bind to the cis-element 5'- ATTAATAACC-3' contained in this in/del sequence. The binding of these transcription factors leads to enhanced upregulation of transcription of the secretory serine protease HTRA1 in transfected cells and AMD patient-derived induced pluripotent stem cells (iPSCs). Overexpression of Htra1 in mice using a CAG-promoter demonstrated increased blood concentration of Htra1 protein, caused upregulation of vascular endothelial growth factor (VEGF), and produced a choroidal neovascularization (CNV)-like phenotype. Finally, a comparison of 478 AMD patients to 481 healthy, age-matched controls from Japan, India, Australia, and the USA showed a statistically increased level of secreted HTRA1 blood concentration in AMD patients compared with age-matched controls. Taken together, these results suggest a common mechanism across ethnicities whereby increased systemic blood circulation of secreted serine protease HTRA1 leads to subsequent degradation of Bruch's membrane and eventual CNV in AMD.


Asunto(s)
Serina Peptidasa A1 que Requiere Temperaturas Altas/genética , Proteínas/genética , Factores de Transcripción TFII/genética , Anciano , Anciano de 80 o más Años , Animales , Femenino , Serina Peptidasa A1 que Requiere Temperaturas Altas/metabolismo , Humanos , Mutación INDEL/genética , Degeneración Macular/genética , Degeneración Macular/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Proteínas/metabolismo , Factores de Transcripción TFII/metabolismo , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-33198939

RESUMEN

We recently reported that cigarette sidestream smoke (CSS) induced inhibition of nucleotide excision repair (NER) and the cause was NER molecule degradation by aldehydes contained in CSS [Carcinogenesis39, 56-65, 2018; Mutat. Res.834, 42-50, 2018]. In this study, we examined the relationship between intracellular glutathione (GSH) levels and CSS-induced NER inhibition. CSS treatment decreased the intracellular GSH level in human keratinocytes HaCaT, in which the repair of pyrimidine (6-4) pyrimidone photoproducts (6-4PPs) after UVB irradiation was suppressed. We used l-buthionine-(S,R)-sulfoximine (BSO) to artificially deplete intracellular GSH level. BSO treatment remarkably accelerated the CSS-induced NER inhibition. The NER inhibition by CSS was attributed to the delay of accumulation of NER molecules (TFIIH and XPG) to DNA damaged sites, which was further enhanced by BSO treatment. CSS degraded TFIIH, and BSO promoted it as expected. Formaldehyde (FA), a major constituent of CSS, showed similar intracellular GSH reduction and NER inhibition, and BSO promoted its inhibitory effect. Five cultured cell lines showed considerable variability in intrinsic GSH levels, and CSS-induced NER inhibitory effect was significantly correlated with the GSH levels. Chemicals like aldehydes are known to react not only with proteins but also with DNA, causing DNA lesions targeted by NER. Our results suggest that the tissues and cells with low intrinsic GSH levels are susceptible to treatment with CSS and electrophilic compounds like aldehydes through NER inhibition, thus leading to higher genotoxicity and carcinogenicity.


Asunto(s)
Aldehídos/farmacología , Reparación del ADN/efectos de los fármacos , Glutatión/metabolismo , Nicotiana/química , Contaminación por Humo de Tabaco/análisis , Butionina Sulfoximina/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Daño del ADN , Glutatión/antagonistas & inhibidores , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Pruebas de Mutagenicidad/métodos , Factores de Transcripción TFIII/metabolismo , Rayos Ultravioleta
12.
Nat Commun ; 11(1): 4905, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32999288

RESUMEN

Transcription factor (TF) IIIC is a conserved eukaryotic six-subunit protein complex with dual function. It serves as a general TF for most RNA polymerase (Pol) III genes by recruiting TFIIIB, but it is also involved in chromatin organization and regulation of Pol II genes through interaction with CTCF and condensin II. Here, we report the structure of the S. cerevisiae TFIIIC subcomplex τA, which contains the most conserved subunits of TFIIIC and is responsible for recruitment of TFIIIB and transcription start site (TSS) selection at Pol III genes. We show that τA binding to its promoter is auto-inhibited by a disordered acidic tail of subunit τ95. We further provide a negative-stain reconstruction of τA bound to the TFIIIB subunits Brf1 and TBP. This shows that a ruler element in τA achieves positioning of TFIIIB upstream of the TSS, and suggests remodeling of the complex during assembly of TFIIIB by TFIIIC.


Asunto(s)
Regulación Fúngica de la Expresión Génica , ARN Polimerasa III/metabolismo , Proteínas de Saccharomyces cerevisiae/ultraestructura , Saccharomyces cerevisiae/genética , Factores de Transcripción TFIII/ultraestructura , Animales , Línea Celular , Microscopía por Crioelectrón , ADN de Hongos/genética , ADN de Hongos/metabolismo , Genes Fúngicos/genética , Insectos , Dominios Proteicos , Multimerización de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Proteínas de Saccharomyces cerevisiae/metabolismo , Factor de Transcripción TFIIIB/genética , Factor de Transcripción TFIIIB/aislamiento & purificación , Factor de Transcripción TFIIIB/metabolismo , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/aislamiento & purificación , Factores de Transcripción TFIII/metabolismo , Sitio de Iniciación de la Transcripción , Iniciación de la Transcripción Genética
13.
Sci Rep ; 10(1): 17440, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33060757

RESUMEN

Transfer RNA genes (tDNAs) are essential genes that encode tRNAs in all species. To understand new functions of tDNAs, other than that of encoding tRNAs, we used ENCODE data to examine binding characteristics of transcription factors (TFs) for all tDNA regions (489 loci) in the human genome. We divided the tDNAs into three groups based on the number of TFs that bound to them. At the two extremes were tDNAs to which many TFs bound (Group 1) and those to which no TFs bound (Group 3). Several TFs involved in chromatin remodeling such as ATF3, EP300 and TBL1XR1 bound to almost all Group 1 tDNAs. Furthermore, almost all Group 1 tDNAs included DNase I hypersensitivity sites and may thus interact with other chromatin regions through their bound TFs, and they showed highly conserved synteny across tetrapods. In contrast, Group 3 tDNAs did not possess these characteristics. These data suggest the presence of a previously uncharacterized function of these tDNAs. We also examined binding of CTCF to tDNAs and their involvement in topologically associating domains (TADs) and lamina-associated domains (LADs), which suggest a new perspective on the evolution and function of tDNAs.


Asunto(s)
Cromatina/química , ARN de Transferencia/metabolismo , Factores de Transcripción/metabolismo , Células A549 , Factor de Transcripción Activador 3/metabolismo , Secuencias de Aminoácidos , Biología Computacional , Bases de Datos Factuales , Proteína p300 Asociada a E1A/metabolismo , Genoma Humano , Células HeLa , Células Hep G2 , Humanos , Células K562 , Dominios Proteicos , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/metabolismo , Sintenía , Factores de Transcripción TFIII/metabolismo
14.
Proc Natl Acad Sci U S A ; 117(35): 21504-21511, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817556

RESUMEN

In fission yeast, the inverted repeats IR-L and IR-R function as boundary elements at the edges of a 20-kb silent heterochromatic domain where nucleosomes are methylated at histone H3K9. Each repeat contains a series of B-box motifs physically associated with the architectural TFIIIC complex and with other factors including the replication regulator Sap1 and the Rix1 complex (RIXC). We demonstrate here the activity of these repeats in heterochromatin formation and maintenance. Deletion of the entire IR-R repeat or, to a lesser degree, deletion of just the B boxes impaired the de novo establishment of the heterochromatic domain. Nucleation proceeded normally at the RNA interference (RNAi)-dependent element cenH but subsequent propagation to the rest of the region occurred at reduced rates in the mutants. Once established, heterochromatin was unstable in the mutants. These defects resulted in bistable populations of cells occupying alternate "on" and "off" epigenetic states. Deleting IR-L in combination with IR-R synergistically tipped the balance toward the derepressed state, revealing a concerted action of the two boundaries at a distance. The nuclear rim protein Amo1 has been proposed to tether the mating-type region and its boundaries to the nuclear envelope, where Amo1 mutants displayed milder phenotypes than boundary mutants. Thus, the boundaries might facilitate heterochromatin propagation and maintenance in ways other than just through Amo1, perhaps by constraining a looped domain through pairing.


Asunto(s)
Proteínas de Unión al ADN/genética , Heterocromatina/metabolismo , Secuencias Invertidas Repetidas/genética , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Silenciador del Gen/fisiología , Heterocromatina/genética , Histonas/metabolismo , Metilación , Proteínas Nucleares/metabolismo , Interferencia de ARN/fisiología , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/metabolismo
15.
Nucleic Acids Res ; 48(20): 11215-11226, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-32747934

RESUMEN

The ChIP-exo assay precisely delineates protein-DNA crosslinking patterns by combining chromatin immunoprecipitation with 5' to 3' exonuclease digestion. Within a regulatory complex, the physical distance of a regulatory protein to DNA affects crosslinking efficiencies. Therefore, the spatial organization of a protein-DNA complex could potentially be inferred by analyzing how crosslinking signatures vary between its subunits. Here, we present a computational framework that aligns ChIP-exo crosslinking patterns from multiple proteins across a set of coordinately bound regulatory regions, and which detects and quantifies protein-DNA crosslinking events within the aligned profiles. By producing consistent measurements of protein-DNA crosslinking strengths across multiple proteins, our approach enables characterization of relative spatial organization within a regulatory complex. Applying our approach to collections of ChIP-exo data, we demonstrate that it can recover aspects of regulatory complex spatial organization at yeast ribosomal protein genes and yeast tRNA genes. We also demonstrate the ability to quantify changes in protein-DNA complex organization across conditions by applying our approach to analyze Drosophila Pol II transcriptional components. Our results suggest that principled analyses of ChIP-exo crosslinking patterns enable inference of spatial organization within protein-DNA complexes.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Proteínas de Unión al ADN/metabolismo , Exonucleasas/química , ARN de Transferencia/genética , Proteínas Ribosómicas/genética , Alineación de Secuencia/métodos , Factores de Transcripción/metabolismo , Algoritmos , Animales , Sitios de Unión , Simulación por Computador , Proteínas de Unión al ADN/química , Bases de Datos Genéticas , Drosophila/química , Drosophila/genética , Drosophila/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN Polimerasa II/química , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa III/química , ARN Polimerasa III/genética , ARN Polimerasa III/metabolismo , ARN de Transferencia/química , ARN de Transferencia/metabolismo , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Análisis de Secuencia de ADN/métodos , Factor de Transcripción TFIIIB/química , Factor de Transcripción TFIIIB/genética , Factor de Transcripción TFIIIB/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción TFIII/química , Factores de Transcripción TFIII/genética , Factores de Transcripción TFIII/metabolismo , Sitio de Iniciación de la Transcripción
16.
J Biol Chem ; 295(14): 4617-4630, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32115405

RESUMEN

Specificity protein 1 (Sp1) is an important transcription factor implicated in numerous cellular processes. However, whether Sp1 is involved in the regulation of RNA polymerase III (Pol III)-directed gene transcription in human cells remains unknown. Here, we first show that filamin A (FLNA) represses Sp1 expression as well as expression of TFIIB-related factor 1 (BRF1) and general transcription factor III C subunit 2 (GTF3C2) in HeLa, 293T, and SaOS2 cell lines stably expressing FLNA-silencing shRNAs. Both BRF1 promoter 4 (BRF1P4) and GTF3C2 promoter 2 (GTF3C2P2) contain putative Sp1-binding sites, suggesting that Sp1 affects Pol III gene transcription by regulating BRF1 and GTF3C2 expression. We demonstrate that Sp1 knockdown inhibits Pol III gene transcription, BRF1 and GTF3C2 expression, and the proliferation of 293T and HeLa cells, whereas Sp1 overexpression enhances these activities. We obtained a comparable result in a cell line in which both FLNA and Sp1 were depleted. These results indicate that Sp1 is involved in the regulation of Pol III gene transcription independently of FLNA expression. Reporter gene assays showed that alteration of Sp1 expression affects BRF1P4 and GTF3C2P2 activation, suggesting that Sp1 modulates Pol III-mediated gene transcription by controlling BRF1 and GTF3C2 gene expression. Further analysis revealed that Sp1 interacts with and thereby promotes the occupancies of TATA box-binding protein, TFIIAα, and p300 at both BRF1P4 and GTF3C2P2. These findings indicate that Sp1 controls Pol III-directed transcription and shed light on how Sp1 regulates cancer cell proliferation.


Asunto(s)
ARN Polimerasa III/metabolismo , Factor de Transcripción Sp1/metabolismo , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factores de Transcripción TFIII/metabolismo , Sitios de Unión , Línea Celular , Proliferación Celular , Proteína p300 Asociada a E1A/metabolismo , Filaminas/antagonistas & inhibidores , Filaminas/genética , Filaminas/metabolismo , Humanos , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Polimerasa III/genética , ARN Interferente Pequeño/metabolismo , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética , Factores Asociados con la Proteína de Unión a TATA/antagonistas & inhibidores , Factores Asociados con la Proteína de Unión a TATA/genética , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Factores de Transcripción TFIII/antagonistas & inhibidores , Factores de Transcripción TFIII/genética , Transcripción Genética , Regulación hacia Arriba
17.
Epigenetics Chromatin ; 13(1): 1, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31918747

RESUMEN

BACKGROUND: Chromatin organization is central to precise control of gene expression. In various eukaryotic species, domains of pervasive cis-chromatin interactions demarcate functional domains of the genomes. In nematode Caenorhabditis elegans, however, pervasive chromatin contact domains are limited to the dosage-compensated sex chromosome, leaving the principle of C. elegans chromatin organization unclear. Transcription factor III C (TFIIIC) is a basal transcription factor complex for RNA polymerase III, and is implicated in chromatin organization. TFIIIC binding without RNA polymerase III co-occupancy, referred to as extra-TFIIIC binding, has been implicated in insulating active and inactive chromatin domains in yeasts, flies, and mammalian cells. Whether extra-TFIIIC sites are present and contribute to chromatin organization in C. elegans remains unknown. RESULTS: We identified 504 TFIIIC-bound sites absent of RNA polymerase III and TATA-binding protein co-occupancy characteristic of extra-TFIIIC sites in C. elegans embryos. Extra-TFIIIC sites constituted half of all identified TFIIIC binding sites in the genome. Extra-TFIIIC sites formed dense clusters in cis. The clusters of extra-TFIIIC sites were highly over-represented within the distal arm domains of the autosomes that presented a high level of heterochromatin-associated histone H3K9 trimethylation (H3K9me3). Furthermore, extra-TFIIIC clusters were embedded in the lamina-associated domains. Despite the heterochromatin environment of extra-TFIIIC sites, the individual clusters of extra-TFIIIC sites were devoid of and resided near the individual H3K9me3-marked regions. CONCLUSION: Clusters of extra-TFIIIC sites were pervasive in the arm domains of C. elegans autosomes, near the outer boundaries of H3K9me3-marked regions. Given the reported activity of extra-TFIIIC sites in heterochromatin insulation in yeasts, our observation raised the possibility that TFIIIC may also demarcate heterochromatin in C. elegans.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Heterocromatina/metabolismo , Factores de Transcripción TFIII/metabolismo , Animales , Sitios de Unión , Caenorhabditis elegans , Heterocromatina/química , Histonas/química , Histonas/metabolismo , Lámina Nuclear/metabolismo , Unión Proteica
18.
Mol Cell Biol ; 40(2)2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31685548

RESUMEN

Interphase chromosomes are organized into topologically associated domains in order to establish and maintain integrity of transcriptional programs that remain poorly understood. Here, we show that condensin II and TFIIIC are recruited to bidirectionally transcribed promoters by a mechanism that is dependent on the retinoblastoma (RB) protein. Long-range chromosome contacts are disrupted by loss of condensin II loading, which leads to altered expression at bidirectional gene pairs. This study demonstrates that mammalian condensin II functions to organize long-range chromosome contacts and regulate transcription at specific genes. In addition, RB dependence of condensin II suggests that widespread misregulation of chromosome contacts and transcriptional alterations are a consequence of RB mutation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cromosomas/metabolismo , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción TFIII/metabolismo , Animales , Células Cultivadas , Cromosomas/genética , Epigénesis Genética , Interfase , Ratones , Regiones Promotoras Genéticas , Activación Transcripcional
19.
Mol Cell ; 77(3): 475-487.e11, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31759822

RESUMEN

How repetitive elements, epigenetic modifications, and architectural proteins interact ensuring proper genome expression remains poorly understood. Here, we report regulatory mechanisms unveiling a central role of Alu elements (AEs) and RNA polymerase III transcription factor C (TFIIIC) in structurally and functionally modulating the genome via chromatin looping and histone acetylation. Upon serum deprivation, a subset of AEs pre-marked by the activity-dependent neuroprotector homeobox Protein (ADNP) and located near cell-cycle genes recruits TFIIIC, which alters their chromatin accessibility by direct acetylation of histone H3 lysine-18 (H3K18). This facilitates the contacts of AEs with distant CTCF sites near promoter of other cell-cycle genes, which also become hyperacetylated at H3K18. These changes ensure basal transcription of cell-cycle genes and are critical for their re-activation upon serum re-exposure. Our study reveals how direct manipulation of the epigenetic state of AEs by a general transcription factor regulates 3D genome folding and expression.


Asunto(s)
Elementos Alu/fisiología , Histonas/metabolismo , Factores de Transcripción TFIII/metabolismo , Acetilación , Elementos Alu/genética , Línea Celular , Cromatina/metabolismo , Cromatina/fisiología , Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Histonas/genética , Proteínas de Homeodominio/genética , Humanos , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional , ARN Polimerasa III/metabolismo , Factores de Transcripción TFIII/genética , Transcripción Genética/genética
20.
Mol Cell Biol ; 39(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30718362

RESUMEN

The genome is packaged and organized in an ordered, nonrandom manner, and specific chromatin segments contact nuclear substructures to mediate this organization. tRNA genes (tDNAs) are binding sites for transcription factors and architectural proteins and are thought to play an important role in the organization of the genome. In this study, we investigate the roles of tDNAs in genomic organization and chromosome function by editing a chromosome so that it lacked any tDNAs. Surprisingly our analyses of this tDNA-less chromosome show that loss of tDNAs does not grossly affect chromatin architecture or chromosome tethering and mobility. However, loss of tDNAs affects local nucleosome positioning and the binding of SMC proteins at these loci. The absence of tDNAs also leads to changes in centromere clustering and a reduction in the frequency of long-range HML-HMR heterochromatin clustering with concomitant effects on gene silencing. We propose that the tDNAs primarily affect local chromatin structure, which results in effects on long-range chromosome architecture.


Asunto(s)
Cromatina/metabolismo , Cromatina/ultraestructura , ARN de Transferencia/genética , Sitios de Unión , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Ensamble y Desensamble de Cromatina , Cromosomas/genética , Cromosomas/metabolismo , Heterocromatina/metabolismo , Heterocromatina/ultraestructura , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción TFIII/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA