Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 508
Filter
1.
Zhonghua Yi Xue Za Zhi ; 104(16): 1410-1417, 2024 Apr 23.
Article in Chinese | MEDLINE | ID: mdl-38644292

ABSTRACT

Objective: To investigate the genetic and expression characteristics of transcription factor IIH (TFIIH) in pre-initiationcomplex in prostate cancer (PCa) and its relationship with prostate cancer progression. Methods: Analyzing the expression characteristics and clinical signification of TFIIH subunits about 495 cases of PCa and 52 cases of adjacent cancer in The Cancer Genome Atlas-Prostate adenocarcinoma (TCGA-PRAD) database. PCa microarray chip was used to verify the correlation between the key factor General Transcription Factor IIH Subunit 4 (GTF2H4) in TFIIH and clinical features. Results: The 495 patients with PCa were (61.01±6.82) years old.The mRNA expression of ERCC3、GTF2H4 and MNAT1 were high in PCa tissues with GS≥8(P<0.05). The expression of GTF2H4 and MNAT1 were relevant to the pathological stages(P<0.05). High expression of GTF2H4 has higher biochemical recurrence (BCR) rate in PCa patients(HR=2.47, 95%CI:1.62-3.77, P<0.001), which has better predictive effect of BCR in PCa patients(The 3rd, 5th, and 7th year AUC all>0.7) than other subunits, and it has been verified in four additional databases. Single-factor Cox regression analysis showed that GTF2H4 were risk factors for BCR (HR=2.470, 95%CI:1.620-3.767, P<0.001) and GTF2H5 were protective factors(HR=0.506,95%CI: 0.336-0.762, P=0.001). The results of immunohistochemical staining showed that the protein expression of GTF2H4 was correlated with the clinical features of PCa patients.The differences of the above results were statistically significant. Conclusion: GTF2H4, the key factor of TFIIH, is highly expressed in PCa and indicates a poor prognosis.


Subject(s)
Computational Biology , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prognosis , Middle Aged , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Aged , Transcription Factors, TFII/metabolism , Transcription Factors, TFII/genetics
2.
Mol Cell ; 84(9): 1699-1710.e6, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38604172

ABSTRACT

The transition from transcription initiation to elongation is highly regulated in human cells but remains incompletely understood at the structural level. In particular, it is unclear how interactions between RNA polymerase II (RNA Pol II) and initiation factors are broken to enable promoter escape. Here, we reconstitute RNA Pol II promoter escape in vitro and determine high-resolution structures of initially transcribing complexes containing 8-, 10-, and 12-nt ordered RNAs and two elongation complexes containing 14-nt RNAs. We suggest that promoter escape occurs in three major steps. First, the growing RNA displaces the B-reader element of the initiation factor TFIIB without evicting TFIIB. Second, the rewinding of the transcription bubble coincides with the eviction of TFIIA, TFIIB, and TBP. Third, the binding of DSIF and NELF facilitates TFIIE and TFIIH dissociation, establishing the paused elongation complex. This three-step model for promoter escape fills a gap in our understanding of the initiation-elongation transition of RNA Pol II transcription.


Subject(s)
Phosphoproteins , Promoter Regions, Genetic , RNA Polymerase II , TATA-Box Binding Protein , Transcription Factor TFIIB , Transcription Factors , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Humans , Transcription Factor TFIIB/metabolism , Transcription Factor TFIIB/genetics , TATA-Box Binding Protein/metabolism , TATA-Box Binding Protein/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Initiation, Genetic , Transcription Factor TFIIH/metabolism , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/chemistry , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Protein Binding , Transcription Factor TFIIA/metabolism , Transcription Factor TFIIA/genetics , Transcription, Genetic , Transcription Elongation, Genetic , RNA/metabolism , RNA/genetics , Transcription Factors, TFII/metabolism , Transcription Factors, TFII/genetics
3.
Hum Genet ; 143(3): 437-453, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38520561

ABSTRACT

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.


Subject(s)
Developmental Disabilities , RNA Polymerase III , Transcription Factors, TFIII , Animals , Child , Child, Preschool , Female , Humans , Male , Alleles , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Intellectual Disability/genetics , Mutation , Pedigree , Phenotype , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , Transcription Factors, TFIII/genetics , Transcription Factors, TFIII/metabolism , Transcription, Genetic , Zebrafish/genetics
4.
Glia ; 72(6): 1117-1135, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38450767

ABSTRACT

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.


Subject(s)
Transcription Factors, TFIII , Transcription Factors, TFII , Williams Syndrome , Mice , Animals , Microglia , Williams Syndrome/genetics , Neurons/metabolism , Disease Models, Animal , Transcription Factors, TFIII/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
5.
Cell Rep ; 43(3): 113867, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38416640

ABSTRACT

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.


Subject(s)
Transcription Factors, TFIII , Transcription Factors, TFII , Williams Syndrome , Humans , Williams Syndrome/genetics , Williams Syndrome/metabolism , Neurons/metabolism , Social Behavior , Phenotype , Transcription Factors, TFIII/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
6.
Curr Opin Struct Biol ; 84: 102766, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38181687

ABSTRACT

RNA polymerase II (Pol II) transcription is regulated by many elongation factors. Among these factors, TFIIF, PAF-RTF1, ELL and Elongin stimulate mRNA chain elongation by Pol II. Cryo-EM structures of Pol II complexes with these elongation factors now reveal some general principles on how elongation factors bind Pol II and how they stimulate transcription. All four elongation factors contact Pol II at domains external 2 and protrusion, whereas TFIIF and ELL additionally bind the Pol II lobe. All factors apparently stabilize cleft-flanking elements, whereas RTF1 and Elongin additionally approach the active site with a latch element and may influence catalysis or translocation. Due to the shared binding sites on Pol II, factor binding is mutually exclusive, and thus it remains to be studied what determines which elongation factors bind at a certain gene and under which condition.


Subject(s)
RNA Polymerase II , Transcription Factors, TFII , RNA Polymerase II/chemistry , Elongin/genetics , Elongin/metabolism , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , Binding Sites , Transcription, Genetic
7.
J Cell Physiol ; 239(3): e31080, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37450667

ABSTRACT

SUMOylation plays an essential role in diverse physiological and pathological processes. Identification of wild-type SUMO1-modification sites by mass spectrometry is still challenging. In this study, we produced a monoclonal SUMO1C-K antibody recognizing SUMOylated peptides and proposed an efficient streamline for identification of SUMOylation sites. We identified 471 SUMOylation sites in 325 proteins from five raw data. These identified sites exhibit a high positive rate when evaluated by mutation-verified SUMOylation sites. We identified many SUMOylated proteins involved in mitochondrial metabolism and non-membrane-bounded organelles formation. We proposed a SUMOylation motif, ΨKXD/EP, where proline is required for efficient SUMOylation. We further revealed SUMOylation of TFII-I was stimulated by growth signals and was required for nucleus-localization of p-ERK1/2. Mutation of SUMOylation sites of TFII-I suppressed tumor cell growth in vitro and in vivo. Taken together, we provided a strategy for personalized identification of wild-type SUMO1-modification sites and revealed the physiological significance of TFII-I SUMOylation in this study.


Subject(s)
Neoplasms , SUMO-1 Protein , Sumoylation , Transcription Factors, TFII , Humans , Antibodies, Monoclonal , Mass Spectrometry , Neoplasms/genetics , Neoplasms/pathology , Peptides/metabolism , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Sumoylation/genetics , Transcription Factors, TFII/metabolism
8.
Cancer Gene Ther ; 30(12): 1702-1714, 2023 12.
Article in English | MEDLINE | ID: mdl-37845349

ABSTRACT

Glioblastoma is the most common malignant tumor in the central nervous system. The general transcription factor IIE subunit beta (GTF2E2) is crucial for physiological and pathological functions, but its roles in the malignant biological function of glioma remain ambiguous. CCK-8, colony formation assays, TUNEL assays, cell migration assays, wound-healing assays, and xenograft model were established to investigate the biological functions of GTF2E2 both in vitro and in vivo. GTF2E2 was overexpressed in glioma and was associated with poor prognosis of glioma patients. Biological functions of GTF2E2 were investigated both in vitro and in vi0vo by multiple experiments. Moreover, we explored the possible mechanisms of GTF2E2. In our results, we demonstrated that GTF2E2 could be regulated by miR-340-5p directly or indirectly. CCND1 was transcriptionally affected by GTF2E2 and glioma progression was then regulated. Our data presented the overexpression of GTF2E2 in glioma and indicated the association between GTF2E2 and glioma prognosis. GTF2E2 was found to be regulated by miR-340-5p and thus affect downstream gene expressions and glioma progression. Our results indicate that GTF2E2 might be a potential target in the diagnosis and treatments of glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , MicroRNAs , Transcription Factors, TFII , Humans , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glioma/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
9.
Front Immunol ; 14: 1067459, 2023.
Article in English | MEDLINE | ID: mdl-36756127

ABSTRACT

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.


Subject(s)
Sjogren's Syndrome , Transcription Factors, TFIII , Transcription Factors, TFII , Humans , Mice , Animals , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin , Protein Binding , Transcription Factors, TFIII/genetics , Transcription Factors, TFIII/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
10.
BMC Genomics ; 23(1): 656, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36114454

ABSTRACT

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.


Subject(s)
Neoplasms, Glandular and Epithelial , RNA, Long Noncoding , Transcription Factors, General , Transcription Factors, TFIII , Transcription Factors, TFII , Gene Expression Regulation, Neoplastic , Humans , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasms, Glandular and Epithelial/genetics , Prognosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Thymus Neoplasms , Transcription Factors, General/genetics , Transcription Factors, General/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , Transcription Factors, TFIII/genetics , Transcription Factors, TFIII/metabolism , Tumor Microenvironment
11.
Hereditas ; 159(1): 23, 2022 May 30.
Article in English | MEDLINE | ID: mdl-35637493

ABSTRACT

BACKGROUND: Hox proteins finely coordinate antero-posterior axis during embryonic development and through their action specific target genes are expressed at the right time and space to determine the embryo body plan. As master transcriptional regulators, Hox proteins recognize DNA through the homeodomain (HD) and interact with a multitude of proteins, including general transcription factors and other cofactors. HD binding specificity increases by protein-protein interactions with a diversity of cofactors that outline the Hox interactome and determine the transcriptional landscape of the selected target genes. All these interactions clearly demonstrate Hox-driven transcriptional regulation, but its precise mechanism remains to be elucidated. RESULTS: Here we report Antennapedia (Antp) Hox protein-protein interaction with the TATA-binding protein (TBP) and the formation of novel trimeric complexes with TFIIEß and Extradenticle (Exd), as well as its participation in transcriptional regulation. Using Bimolecular Fluorescence Complementation (BiFC), we detected the interaction of Antp-TBP and, in combination with Förster Resonance Energy Transfer (BiFC-FRET), the formation of the trimeric complex with TFIIEß and Exd in living cells. Mutational analysis showed that Antp interacts with TBP through their N-terminal polyglutamine-stretches. The trimeric complexes of Antp-TBP with TFIIEß and Exd were validated using different Antp mutations to disrupt the trimeric complexes. Interestingly, the trimeric complex Antp-TBP-TFIIEß significantly increased the transcriptional activity of Antp, whereas Exd diminished its transactivation. CONCLUSIONS: Our findings provide important insights into the Antp interactome with the direct interaction of Antp with TBP and the two new trimeric complexes with TFIIEß and Exd. These novel interactions open the possibility to analyze promoter function and gene expression to measure transcription factor binding dynamics at target sites throughout the genome.


Subject(s)
Antennapedia Homeodomain Protein , Drosophila Proteins , Homeodomain Proteins , TATA-Box Binding Protein , Transcription Factors, TFII , Transcription Factors , Antennapedia Homeodomain Protein/genetics , Antennapedia Homeodomain Protein/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
12.
J Biol Chem ; 298(6): 101963, 2022 06.
Article in English | MEDLINE | ID: mdl-35452682

ABSTRACT

Formation of transcription factor (TF)-coregulator complexes is a key step in transcriptional regulation, with coregulators having essential functions as hub nodes in molecular networks. How specificity and selectivity are maintained in these nodes remain open questions. In this work, we addressed specificity in transcriptional networks using complexes formed between TFs and αα-hubs, which are defined by a common αα-hairpin secondary structure motif, as a model. Using NMR spectroscopy and binding thermodynamics, we analyzed the structure, dynamics, stability, and ligand-binding properties of the Arabidopsis thaliana RST domains from TAF4 and known binding partner RCD1, and the TAFH domain from human TAF4, allowing comparison across species, functions, and architectural contexts. While these αα-hubs shared the αα-hairpin motif, they differed in length and orientation of accessory helices as well as in their thermodynamic profiles of ligand binding. Whereas biologically relevant RCD1-ligand pairs displayed high affinity driven by enthalpy, TAF4-ligand interactions were entropy driven and exhibited less binding-induced structuring. We in addition identified a thermal unfolding state with a structured core for all three domains, although the temperature sensitivity differed. Thermal stability studies suggested that initial unfolding of the RCD1-RST domain localized around helix 1, lending this region structural malleability, while effects in TAF4-RST were more stochastic, suggesting variability in structural adaptability upon binding. Collectively, our results support a model in which hub structure, flexibility, and binding thermodynamics contribute to αα-hub-TF binding specificity, a finding of general relevance to the understanding of coregulator-ligand interactions and interactome sizes.


Subject(s)
Arabidopsis Proteins/chemistry , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry , Transcription Factors, TFII/chemistry , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Humans , Ligands , Nuclear Proteins/metabolism , Protein Binding , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism , Transcription Factors, TFII/metabolism
13.
Mol Cell ; 82(3): 660-676.e9, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35051353

ABSTRACT

Previous structural studies of the initiation-elongation transition of RNA polymerase II (pol II) transcription have relied on the use of synthetic oligonucleotides, often artificially discontinuous to capture pol II in the initiating state. Here, we report multiple structures of initiation complexes converted de novo from a 33-subunit yeast pre-initiation complex (PIC) through catalytic activities and subsequently stalled at different template positions. We determine that PICs in the initially transcribing complex (ITC) can synthesize a transcript of ∼26 nucleotides before transitioning to an elongation complex (EC) as determined by the loss of general transcription factors (GTFs). Unexpectedly, transition to an EC was greatly accelerated when an ITC encountered a downstream EC stalled at promoter proximal regions and resulted in a collided head-to-end dimeric EC complex. Our structural analysis reveals a dynamic state of TFIIH, the largest of GTFs, in PIC/ITC with distinct functional consequences at multiple steps on the pathway to elongation.


Subject(s)
RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Transcription Initiation, Genetic , Cryoelectron Microscopy , Gene Expression Regulation, Fungal , Models, Molecular , Promoter Regions, Genetic , Protein Conformation , RNA Polymerase II/genetics , RNA Polymerase II/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Structure-Activity Relationship , Time Factors , Transcription Elongation, Genetic , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
14.
Nat Genet ; 54(2): 180-193, 2022 02.
Article in English | MEDLINE | ID: mdl-35039641

ABSTRACT

How gene expression is controlled to preserve human T cell quiescence is poorly understood. Here we show that non-canonical splicing variants containing long interspersed nuclear element 1 (LINE1) enforce naive CD4+ T cell quiescence. LINE1-containing transcripts are derived from CD4+ T cell-specific genes upregulated during T cell activation. In naive CD4+ T cells, LINE1-containing transcripts are regulated by the transcription factor IRF4 and kept at chromatin by nucleolin; these transcripts act in cis, hampering levels of histone 3 (H3) lysine 36 trimethyl (H3K36me3) and stalling gene expression. T cell activation induces LINE1-containing transcript downregulation by the splicing suppressor PTBP1 and promotes expression of the corresponding protein-coding genes by the elongating factor GTF2F1 through mTORC1. Dysfunctional T cells, exhausted in vitro or tumor-infiltrating lymphocytes (TILs), accumulate LINE1-containing transcripts at chromatin. Remarkably, depletion of LINE1-containing transcripts restores TIL effector function. Our study identifies a role for LINE1 elements in maintaining T cell quiescence and suggests that an abundance of LINE1-containing transcripts is critical for T cell effector function and exhaustion.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Chromatin/metabolism , Gene Expression Regulation , Long Interspersed Nucleotide Elements , RNA Splicing , CD4-Positive T-Lymphocytes/immunology , Chromatin/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Histones/metabolism , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphoproteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors, TFII/metabolism , Transcription, Genetic , Nucleolin
15.
Cells ; 10(12)2021 12 12.
Article in English | MEDLINE | ID: mdl-34944018

ABSTRACT

Mayaro virus (MAYV) is an emerging mosquito-transmitted virus that belongs to the genus Alphavirus within the family Togaviridae. Humans infected with MAYV often develop chronic and debilitating arthralgia and myalgia. The virus is primarily maintained via a sylvatic cycle, but it has the potential to adapt to urban settings, which could lead to large outbreaks. The interferon (IFN) system is a critical antiviral response that limits replication and pathogenesis of many different RNA viruses, including alphaviruses. Here, we investigated how MAYV infection affects the induction phase of the IFN response. Production of type I and III IFNs was efficiently suppressed during MAYV infection, and mapping revealed that expression of the viral non-structural protein 2 (nsP2) was sufficient for this process. Interactome analysis showed that nsP2 interacts with DNA-directed RNA polymerase II subunit A (Rpb1) and transcription initiation factor IIE subunit 2 (TFIIE2), which are host proteins required for RNA polymerase II-mediated transcription. Levels of these host proteins were reduced by nsP2 expression and during infection by MAYV and related alphaviruses, suggesting that nsP2-mediated inhibition of host cell transcription is an important aspect of how some alphaviruses block IFN induction. The findings from this study may prove useful in design of vaccines and antivirals, which are currently not available for protection against MAYV and infection by other alphaviruses.


Subject(s)
Alphavirus/metabolism , Host-Pathogen Interactions , Interferons/metabolism , Protein Subunits/metabolism , Transcription Factors, TFII/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Down-Regulation , Humans , Interferon Regulatory Factor-3/metabolism , Protein Binding , Protein Transport , RNA Polymerase II/metabolism , Transcription, Genetic
16.
J Genet ; 1002021.
Article in English | MEDLINE | ID: mdl-34553694

ABSTRACT

MicroRNAs play important roles in the pathogenesis of human diseases by regulating target gene expression in specific cells or tissues. Previously, we identified microRNA 452 (MIR452), which was specifically upregulated in early stage of human colorectal cancer (CRC) tissues. Here, we show the biological role of MIR452 and general transcription factor IIE subunit 1 (GTF2E1) in colorectal cancer. A luciferase reporter system was used to confirm the effect of MIR452 on GTF2E1 expression. The expression levels of MIR452 and the target genes were evaluated by quantitative RT-PCR (qRT-PCR) and western blotting. We verified the association between MIR452 and the GTF2E1 expression and found that GTF2E1 transcripts were directly downregulated by MIR452. The mRNA and protein levels of GTF2E1 were also downregulated in CRC cells upon transfection with MIR452. GTF2E1 protein expression was decreased in CRC tissues compared to adjacent nontumour tissues. These results suggest that MIR452 might directly or indirectly regulate the genes transcription related to CRC by downregulating GTF2E1 expression.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Transcription Factors, TFII/genetics , Base Sequence , Cell Line, Tumor , Down-Regulation/genetics , Female , Humans , Male , MicroRNAs/genetics , Middle Aged , Transcription Factors, TFII/metabolism
17.
J Phys Chem B ; 125(39): 10917-10927, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34550709

ABSTRACT

Intrinsically disordered protein (IDP) sequences often contain a high proportion of charged residues in conjunction with their high degree of hydrophilicity and solvation. For high net charge IDPs, long-range electrostatic interactions are thought to play a role in modulating the strength or kinetics of protein-protein interactions. In this work, we examined intramolecular interactions mediated by charged regions of a model IDP, the C-terminal tail of the phosphatase Fcp1. Specifically, this work focuses on intermolecular interactions between acidic and basic patches in the primary structure of Fcp1 and their contributions to binding its predominantly basic partner, the winged helix domain of Rap74. We observe both intramolecular and intermolecular interactions through paramagnetic relaxation enhancement (PRE) consistent with oppositely charged regions associating with one another, both in unbound Fcp1 and in the Fcp1-Rap74 complex. Formation of this complex is strongly driven by hydrophobic interactions in the minimal binding motif. Here, we test the hypothesis that charged residues in Fcp1 that flank the binding helix also contribute to the strength of binding. Charge inversion mutations in Fcp1 generally support this hypothesis, while PRE data suggest substitution of observed transient interactions in the unbound ensemble for similarly transient interactions with Rap74 in the complex.


Subject(s)
Transcription Factors, TFII , Phosphoprotein Phosphatases/metabolism , Protein Binding , Protein Structure, Secondary , Static Electricity , Transcription Factors, TFII/metabolism
18.
Biomolecules ; 11(8)2021 07 23.
Article in English | MEDLINE | ID: mdl-34439758

ABSTRACT

BACKGROUND: Aspirin-exacerbated respiratory disease (AERD) is a syndrome characterised by chronic rhinosinusitis, nasal polyps, asthma and aspirin intolerance. An imbalance of eicosanoid metabolism with anover-production of cysteinyl leukotrienes (CysLTs) has been associated with AERD. However, the precise mechanisms underlying AERD are unknown. OBJECTIVE: To establish the transcriptome of the nasal polyp airway epithelial cells derived from AERD patients to discover gene expression patterns in this disease. METHODS: Nasal airway epithelial cells were isolated from 12 AERD polyps and 8 AERD non-polyp nasal mucosa samples as controls from the same subjects. Utilising the Illumina HiSeq 2500 platform, RNA samples were sequenced. Potential gene candidate DMRT3 was selected from the differentially-expressed genes for validation. RESULTS: Comparative transcriptome profiling of nasal epithelial cells was accomplished in AERD. A total of 20 genes had twofold mean regulation expression differences or greater. In addition, 8 genes were upregulated, including doublesex and mab-3 related transcription factor 3 (DMRT3), and 12 genes were downregulated. Differentially regulated genes comprised roles in inflammation, defence and immunity. Metabolic process and embryonic development pathways were significantly enriched. Enzyme-linked immune sorbent assay (ELISA) results of DMRT3 in AERD patients were significantly upregulated compared to controls (p = 0.03). Immunohistochemistry (IHC) of AERD nasal polyps localised DMRT3 and was predominantly released in the airway epithelia. CONCLUSION: Findings suggest that DMRT3 could be potentially involved in nasal polyp development in AERD patients. Furthermore, several genes are downregulated, hinting at the dedifferentiation phenomenon in AERD polyps. However, further studies are imperative to confirm the exact mechanism of polyp formation in AERD patients.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Nasal Polyps/metabolism , Respiration Disorders/drug therapy , Respiration Disorders/metabolism , Transcription Factors, TFII/metabolism , Transcriptome , Adult , Aspirin/adverse effects , Asthma, Aspirin-Induced/genetics , Asthma, Aspirin-Induced/metabolism , Chronic Disease , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Leukotrienes/metabolism , Male , Middle Aged , Nasal Lavage , Nasal Polyps/immunology , RNA-Seq , Sinusitis/immunology , Sinusitis/metabolism , Skin Tests
19.
Mol Cell ; 81(17): 3576-3588.e6, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34384542

ABSTRACT

RNA polymerase II (RNA Pol II) transcription reconstituted from purified factors suggests pre-initiation complexes (PICs) can assemble by sequential incorporation of factors at the TATA box. However, these basal transcription reactions are generally independent of activators and co-activators. To study PIC assembly under more realistic conditions, we used single-molecule microscopy to visualize factor dynamics during activator-dependent reactions in nuclear extracts. Surprisingly, RNA Pol II, TFIIF, and TFIIE can pre-assemble on enhancer-bound activators before loading into PICs, and multiple RNA Pol II complexes can bind simultaneously to create a localized cluster. Unlike TFIIF and TFIIE, TFIIH binding is singular and dependent on the basal promoter. Activator-tethered factors exhibit dwell times on the order of seconds. In contrast, PICs can persist on the order of minutes in the absence of nucleotide triphosphates, although TFIIE remains unexpectedly dynamic even after TFIIH incorporation. Our kinetic measurements lead to a new branched model for activator-dependent PIC assembly.


Subject(s)
Mediator Complex/metabolism , RNA Polymerase II/metabolism , Transcription Initiation, Genetic/physiology , Cell Nucleus/metabolism , Mediator Complex/genetics , Promoter Regions, Genetic , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Single Molecule Imaging , TATA Box/genetics , TATA-Box Binding Protein/genetics , Transcription Factor TFIIH/metabolism , Transcription Factors, TFII/metabolism , Transcription, Genetic/genetics
20.
DNA Repair (Amst) ; 106: 103175, 2021 10.
Article in English | MEDLINE | ID: mdl-34280590

ABSTRACT

The multifunctional transcription factor TFII-I, encoded by the GTF2I gene, is implicated in various biological pathways, and associated with multiple human disorders. Evidence is also mounting to suggest that TFII-I is involved in DNA damage repair pathways. Here I bring together these recent observations and suggest a connection between transcriptional and DNA repair functions of TFII-I.


Subject(s)
DNA Damage , DNA Repair , Transcription Factors, TFII/metabolism , Animals , DNA/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...