Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Publication year range
1.
Mol Cell ; 71(6): 911-922.e4, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30122535

ABSTRACT

NusG/Spt5 proteins are the only transcription factors utilized by all cellular organisms. In enterobacteria, NusG antagonizes the transcription termination activity of Rho, a hexameric helicase, during the synthesis of ribosomal and actively translated mRNAs. Paradoxically, NusG helps Rho act on untranslated transcripts, including non-canonical antisense RNAs and those arising from translational stress; how NusG fulfills these disparate functions is unknown. Here, we demonstrate that NusG activates Rho by assisting helicase isomerization from an open-ring, RNA-loading state to a closed-ring, catalytically active translocase. A crystal structure of closed-ring Rho in complex with NusG reveals the physical basis for this activation and further explains how Rho is excluded from translationally competent RNAs. This study demonstrates how a universally conserved transcription factor acts to modulate the activity of a ring-shaped ATPase motor and establishes how the innate sequence bias of a termination factor can be modulated to silence pervasive, aberrant transcription.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Escherichia coli Proteins/physiology , Peptide Elongation Factors/physiology , Transcription Factors/physiology , Transcription Termination, Genetic/physiology , Transcriptional Elongation Factors/physiology , Bacterial Proteins , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Models, Molecular , Peptide Elongation Factors/metabolism , Protein Conformation , RNA, Bacterial , Rho Factor/metabolism , Rho Factor/physiology , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/physiology
2.
Mol Cell ; 72(4): 687-699.e6, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30318445

ABSTRACT

Spt6 is a conserved factor that controls transcription and chromatin structure across the genome. Although Spt6 is viewed as an elongation factor, spt6 mutations in Saccharomyces cerevisiae allow elevated levels of transcripts from within coding regions, suggesting that Spt6 also controls initiation. To address the requirements for Spt6 in transcription and chromatin structure, we have combined four genome-wide approaches. Our results demonstrate that Spt6 represses transcription initiation at thousands of intragenic promoters. We characterize these intragenic promoters and find sequence features conserved with genic promoters. Finally, we show that Spt6 also regulates transcription initiation at most genic promoters and propose a model of initiation site competition to account for this. Together, our results demonstrate that Spt6 controls the fidelity of transcription initiation throughout the genome.


Subject(s)
Histone Chaperones/genetics , Histone Chaperones/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Transcription Initiation, Genetic/physiology , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/physiology , Chromatin/physiology , Gene Expression Regulation, Fungal/genetics , Histone Chaperones/metabolism , Histones/physiology , Nuclear Proteins , Nucleosomes , Peptide Elongation Factors/physiology , Promoter Regions, Genetic/genetics , RNA Polymerase II , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/physiology , Transcription Factors/physiology , Transcription Initiation Site/physiology , Transcription, Genetic/genetics , Transcriptional Elongation Factors/metabolism
3.
Genes Dev ; 32(1): 26-41, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29378787

ABSTRACT

Regulation by gene-distal enhancers is critical for cell type-specific and condition-specific patterns of gene expression. Thus, to understand the basis of gene activity in a given cell type or tissue, we must identify the precise locations of enhancers and functionally characterize their behaviors. Here, we demonstrate that transcription is a nearly universal feature of enhancers in Drosophila and mammalian cells and that nascent RNA sequencing strategies are optimal for identification of both enhancers and superenhancers. We dissect the mechanisms governing enhancer transcription and discover remarkable similarities to transcription at protein-coding genes. We show that RNA polymerase II (RNAPII) undergoes regulated pausing and release at enhancers. However, as compared with mRNA genes, RNAPII at enhancers is less stable and more prone to early termination. Furthermore, we found that the level of histone H3 Lys4 (H3K4) methylation at enhancers corresponds to transcriptional activity such that highly active enhancers display H3K4 trimethylation rather than the H3K4 monomethylation considered a hallmark of enhancers. Finally, our work provides insights into the unique characteristics of superenhancers, which stimulate high-level gene expression through rapid pause release; interestingly, this property renders associated genes resistant to the loss of factors that stabilize paused RNAPII.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Transcription Elongation, Genetic , Animals , Chromatin/chemistry , Chromosomal Proteins, Non-Histone/physiology , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/physiology , Embryonic Stem Cells/metabolism , Histones/metabolism , Mice , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA, Untranslated/biosynthesis , Transcription Initiation Site , Transcription, Genetic , Transcriptional Elongation Factors/physiology
4.
Nucleic Acids Res ; 49(9): 4944-4953, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33877330

ABSTRACT

Transcription elongation can be affected by numerous types of obstacles, such as nucleosome, pausing sequences, DNA lesions and non-B-form DNA structures. Spt4/5 and Elf1 are conserved transcription elongation factors that promote RNA polymerase II (Pol II) bypass of nucleosome and pausing sequences. Importantly, genetic studies have shown that Spt4/5 plays essential roles in the transcription of expanded nucleotide repeat genes associated with inherited neurological diseases. Here, we investigate the function of Spt4/5 and Elf1 in the transcription elongation of CTG•CAG repeat using an in vitro reconstituted yeast transcription system. We found that Spt4/5 helps Pol II transcribe through the CTG•CAG tract duplex DNA, which is in good agreement with its canonical roles in stimulating transcription elongation. In sharp contrast, surprisingly, we revealed that Spt4/5 greatly inhibits Pol II transcriptional bypass of CTG and CAG slip-out structures. Furthermore, we demonstrated that transcription elongation factor Elf1 individually and cooperatively with Spt4/5 inhibits Pol II bypass of the slip-out structures. This study uncovers the important functional interplays between template DNA structures and the function of transcription elongation factors. This study also expands our understanding of the functions of Spt4/5 and Elf1 in transcriptional processing of trinucleotide repeat DNA.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , DNA, B-Form/chemistry , DNA/chemistry , Nuclear Proteins/physiology , RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins/physiology , Transcription, Genetic , Transcriptional Elongation Factors/physiology , Trinucleotide Repeats
5.
Mol Biol Rep ; 49(2): 1303-1320, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34807377

ABSTRACT

BACKGROUND: Transcription elongation is a dynamic and tightly regulated step of gene expression in eukaryotic cells. Eleven nineteen Lysine rich Leukemia (ELL) and ELL Associated Factors (EAF) family of conserved proteins are required for efficient RNA polymerase II-mediated transcription elongation. Orthologs of these proteins have been identified in different organisms, including fission yeast and humans. METHODS AND RESULTS: In the present study, we have examined the sequence, structural and functional conservation between the fission yeast and human ELL and EAF orthologs. Our computational analysis revealed that these proteins share some sequence characteristics, and were predominantly disordered in both organisms. Our functional complementation assays revealed that both human ELL and EAF proteins could complement the lack of ell1+ or eaf1+ in Schizosaccharomyces pombe respectively. Furthermore, our domain mapping experiments demonstrated that both the amino and carboxyl terminal domains of human EAF proteins could functionally complement the S. pombe eaf1 deletion phenotypes. However, only the carboxyl-terminus domain of human ELL was able to partially rescue the phenotypes associated with lack of ell1+ in S. pombe. CONCLUSIONS: Collectively, our work adds ELL-EAF to the increasing list of human-yeast complementation gene pairs, wherein the simpler fission yeast can be used to further enhance our understanding of the role of these proteins in transcription elongation and human disease.


Subject(s)
Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , Amino Acid Sequence/genetics , Humans , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , RNA Polymerase II/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Sequence Homology , Transcription Factors/genetics , Transcription, Genetic/genetics , Transcription, Genetic/physiology , Transcriptional Elongation Factors/physiology
6.
Exp Cell Res ; 399(2): 112445, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33417923

ABSTRACT

Melanoma is characterized by high mortality and poor prognosis due to metastasis. AFF4 (AF4/FMR2 family member 4), as a scaffold protein, is a component of the super elongation complex (SEC), and is involved in the progression of tumors, e.g., leukemia, head and neck squamous cell carcinoma (HNSCC). However, few studies on AFF4 have focused on melanoma. Here, AFF4 expression levels and clinicopathological features were evaluated in melanoma tissue samples. Then, we performed cell proliferation, migration and invasion assays in A375 and A2058 cells lines in vitro to evaluate the role of AFF4 in melanoma. The effects of AFF4 knockdown in vivo were characterized via a xenograft mouse model. Finally, the correlation between c-Jun and AFF4 protein levels in melanoma was analyzed by rescue assay and immunohistochemistry (IHC). We found that AFF4 expression was upregulated in melanoma tumor tissues and that AFF4 protein expression was also closely related to the prognosis of patients with cutaneous melanoma. Moreover, AFF4 could promote the invasion and migration of melanoma cells by mediating epithelial to mesenchymal transition (EMT). AFF4 might regulate c-Jun activity to promote the invasion and migration of melanoma cells. Importantly, c-Jun was regulated by the AFF4 promoted melanoma tumorigenesis in vivo. Taken together, AFF4 may be a novel oncogene that promotes melanoma progression through regulation of c-Jun activity.


Subject(s)
Melanoma/pathology , Proto-Oncogene Proteins c-jun/genetics , Skin Neoplasms/pathology , Transcriptional Elongation Factors/physiology , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Melanoma/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-jun/metabolism , Skin Neoplasms/genetics
7.
Proc Natl Acad Sci U S A ; 116(44): 22140-22151, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611376

ABSTRACT

Soon after exposure to genotoxic reagents, mammalian cells inhibit transcription to prevent collisions with repair machinery and to mount a proper DNA damage response. However, mechanisms underlying early transcriptional inhibition are poorly understood. In this report, we show that site-specific acetylation of super elongation complex (SEC) subunit AFF1 by p300 reduces its interaction with other SEC components and impairs P-TEFb-mediated C-terminal domain phosphorylation of RNA polymerase II both in vitro and in vivo. Reexpression of wild-type AFF1, but not an acetylation mimic mutant, restores SEC component recruitment and target gene expression in AFF1 knockdown cells. Physiologically, we show that, upon genotoxic exposure, p300-mediated AFF1 acetylation is dynamic and strongly correlated with concomitant global down-regulation of transcription-and that this can be reversed by overexpression of an acetylation-defective AFF1 mutant. Therefore, we describe a mechanism of dynamic transcriptional regulation involving p300-mediated acetylation of a key elongation factor during genotoxic stress.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein/metabolism , Transcriptional Elongation Factors/metabolism , Acetylation , DNA Repair , DNA-Binding Proteins/physiology , Genomic Instability , Humans , Phosphorylation , RNA Polymerase II/metabolism , Stress, Physiological , Transcription, Genetic , Transcriptional Elongation Factors/physiology
8.
Annu Rev Genet ; 47: 483-508, 2013.
Article in English | MEDLINE | ID: mdl-24050178

ABSTRACT

Elongation is becoming increasingly recognized as a critical step in eukaryotic transcriptional regulation. Although traditional genetic and biochemical studies have identified major players of transcriptional elongation, our understanding of the importance and roles of these factors is evolving rapidly through the recent advances in genome-wide and single-molecule technologies. Here, we focus on how elongation can modulate the transcriptional outcome through the rate-liming step of RNA polymerase II (Pol II) pausing near promoters and how the participating factors were identified. Among the factors we describe are the pausing factors--NELF (negative elongation factor) and DSIF (DRB sensitivity-inducing factor)--and P-TEFb (positive elongation factor b), which is the key player in pause release. We also describe the high-resolution view of Pol II pausing and propose nonexclusive models for how pausing is achieved. We then discuss Pol II elongation through the bodies of genes and the roles of FACT and SPT6, factors that allow Pol II to move through nucleosomes.


Subject(s)
Transcription Elongation, Genetic/physiology , Animals , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Eukaryotic Cells/metabolism , High Mobility Group Proteins/physiology , Humans , Mammals/genetics , Models, Genetic , Nucleosomes/genetics , Phosphorylation , Prokaryotic Cells/metabolism , Promoter Regions, Genetic/genetics , Protein Interaction Mapping , RNA Caps/genetics , RNA Polymerase II/genetics , RNA Splicing , Ribosomes/genetics , Transcription Factors/physiology , Transcriptional Elongation Factors/physiology
9.
RNA ; 25(10): 1298-1310, 2019 10.
Article in English | MEDLINE | ID: mdl-31289129

ABSTRACT

There is increasing evidence from yeast to humans that pre-mRNA splicing occurs mainly cotranscriptionally, such that splicing and transcription are functionally coupled. Currently, there is little insight into the contribution of the core transcription elongation machinery to cotranscriptional spliceosome assembly and pre-mRNA splicing. Spt5 is a member of the core transcription elongation machinery and an essential protein, whose absence in budding yeast causes defects in pre-mRNA splicing. To determine how Spt5 affects pre-mRNA splicing, we used the auxin-inducible degron system to conditionally deplete Spt5 in Saccharomyces cerevisiae and assayed effects on cotranscriptional spliceosome assembly and splicing. We show that Spt5 is needed for efficient splicing and for the accumulation of U5 snRNPs at intron-containing genes, and therefore for stable cotranscriptional assembly of spliceosomes. The defect in cotranscriptional spliceosome assembly can explain the relatively mild splicing defect as being a consequence of the failure of cotranscriptional splicing. Coimmunoprecipitation of Spt5 with core spliceosomal proteins and all spliceosomal snRNAs suggests a model whereby Spt5 promotes cotranscriptional pre-mRNA splicing by stabilizing the association of U5 snRNP with spliceosome complexes as they assemble on the nascent transcript. If this phenomenon is conserved in higher eukaryotes, it has the potential to be important for cotranscriptional regulation of alternative splicing.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Saccharomyces cerevisiae/metabolism , Spliceosomes , Transcription, Genetic , Transcriptional Elongation Factors/physiology , Chromosomal Proteins, Non-Histone/metabolism , Immunoprecipitation , Protein Binding , RNA Splicing , Ribonucleoprotein, U5 Small Nuclear/genetics , Transcriptional Elongation Factors/metabolism
10.
Nucleic Acids Res ; 47(12): 6488-6503, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31020314

ABSTRACT

Ribosomal RNA synthesis in Escherichia coli involves a transcription complex, in which RNA polymerase is modified by a signal element on the transcript, Nus factors A, B, E and G, ribosomal protein S4 and inositol mono-phosphatase SuhB. This complex is resistant to ρ-dependent termination and facilitates ribosomal RNA folding, maturation and subunit assembly. The functional contributions of SuhB and their structural bases are presently unclear. We show that SuhB directly binds the RNA signal element and the C-terminal AR2 domain of NusA, and we delineate the atomic basis of the latter interaction by macromolecular crystallography. SuhB recruitment to a ribosomal RNA transcription complex depends on the RNA signal element but not on the NusA AR2 domain. SuhB in turn is required for stable integration of the NusB/E dimer into the complex. In vitro transcription assays revealed that SuhB is crucial for delaying or suppressing ρ-dependent termination, that SuhB also can reduce intrinsic termination, and that SuhB-AR2 contacts contribute to these effects. Together, our results reveal functions of SuhB during ribosomal RNA synthesis and delineate some of the underlying molecular interactions.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/genetics , Phosphoric Monoester Hydrolases/chemistry , RNA, Ribosomal/biosynthesis , Transcription Factors/chemistry , Transcriptional Elongation Factors/chemistry , Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/physiology , Models, Molecular , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/physiology , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Transcription, Genetic , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/physiology
11.
Gut ; 69(2): 329-342, 2020 02.
Article in English | MEDLINE | ID: mdl-31439637

ABSTRACT

OBJECTIVE: Facilitates Chromatin Transcription (FACT) complex is a histone chaperone participating in DNA repair-related and transcription-related chromatin dynamics. In this study, we investigated its oncogenic functions, underlying mechanisms and therapeutic implications in human hepatocellular carcinoma (HCC). DESIGN: We obtained HCC and its corresponding non-tumorous liver samples from 16 patients and identified FACT complex as the most upregulated histone chaperone by RNA-Seq. We further used CRISPR-based gene activation and knockout systems to demonstrate the functions of FACT complex in HCC growth and metastasis. Functional roles and mechanistic insights of FACT complex in oxidative stress response were investigated by ChIP assay, flow cytometry, gene expression assays and 4sU-DRB transcription elongation assay. Therapeutic effect of FACT complex inhibitor, Curaxin, was tested in both in vitro and in vivo models. RESULTS: We showed that FACT complex was remarkably upregulated in HCC and contributed to HCC progression. Importantly, we unprecedentedly revealed an indispensable role of FACT complex in NRF2-driven oxidative stress response. Oxidative stress prevented NRF2 and FACT complex from KEAP1-mediated protein ubiquitination and degradation. Stabilised NRF2 and FACT complex form a positive feedback loop; NRF2 transcriptionally activates the FACT complex, while FACT complex promotes the transcription elongation of NRF2 and its downstream antioxidant genes through facilitating rapid nucleosome disassembly for the passage of RNA polymerase. Therapeutically, Curaxin effectively suppressed HCC growth and sensitised HCC cell to sorafenib. CONCLUSION: In conclusion, our findings demonstrated that FACT complex is essential for the expeditious HCC oxidative stress response and is a potential therapeutic target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Histone Chaperones/physiology , Liver Neoplasms/physiopathology , Oxidative Stress/physiology , Transcriptional Elongation Factors/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carbazoles/pharmacology , Carbazoles/therapeutic use , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Gene Knockout Techniques/methods , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/biosynthesis , High Mobility Group Proteins/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/physiopathology , Liver Neoplasms, Experimental/prevention & control , Mice, Inbred BALB C , Mice, Nude , Oxidative Stress/genetics , Sorafenib/pharmacology , Sorafenib/therapeutic use , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology , Transcriptional Elongation Factors/antagonists & inhibitors , Transcriptional Elongation Factors/biosynthesis , Transcriptional Elongation Factors/genetics , Up-Regulation/physiology , Xenograft Model Antitumor Assays
12.
Nucleic Acids Res ; 45(11): 6362-6374, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28379497

ABSTRACT

RNA polymerase II (RNAPII) passes through the nucleosome in a coordinated manner, generating several intermediate nucleosomal states as it breaks and then reforms histone-DNA contacts ahead of and behind it, respectively. Several studies have defined transcription-induced nucleosome intermediates using only RNA Polymerase. However, RNAPII is decorated with elongation factors as it transcribes the genome. One such factor, Spt4/5, becomes an integral component of the elongation complex, making direct contact with the 'jaws' of RNAPII and nucleic acids in the transcription scaffold. We have characterized the effect of incorporating Spt4/5 into the elongation complex on transcription through the 601R nucleosome. Spt4/5 suppressed RNAPII pausing at the major H3/H4-induced arrest point, resulting in downstream re-positioning of RNAPII further into the nucleosome. Using a novel single molecule FRET system, we found that Spt4/5 affected the kinetics of DNA re-wrapping and stabilized a nucleosomal intermediate with partially unwrapped DNA behind RNAPII. Comparison of nucleosomes of different sequence polarities suggest that the strength of the DNA-histone interactions behind RNAPII specifies the Spt4/5 requirement. We propose that Spt4/5 may be important to coordinate the mechanical movement of RNAPII through the nucleosome with co-transcriptional chromatin modifications during transcription, which is affected by the strength of histone-DNA interactions.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Nuclear Proteins/physiology , RNA Polymerase II/genetics , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/enzymology , Transcriptional Elongation Factors/physiology , DNA, Fungal/physiology , Gene Expression Regulation, Fungal , Nucleosomes/physiology , Protein Binding , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Transcription, Genetic
13.
RNA ; 22(4): 571-82, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26873599

ABSTRACT

Coupling between transcription and RNA processing is key for gene regulation. Using live-cell photobleaching techniques, we investigated the factor TCERG1, which coordinates transcriptional elongation with splicing. We demonstrate that TCERG1 is highly mobile in the nucleoplasm and that this mobility is slightly decreased when it is associated with speckles. Dichloro-1-ß-D-ribofuranosylbenzimidazole (DRB) but not α-amanitin treatment reduced the mobility of TCERG1, which suggests interaction with paused transcription elongation complexes. We found that TCERG1 mobility is rapid at the transcription site (TS) of a reporter that splices post-transcriptionally and that TCERG1 is recruited to the active TS independent of the CTD of RNAPII, thus excluding phosphorylated CTD as a requirement for recruiting this factor to the TS. Importantly, the mobility of TCERG1 is reduced when the reporter splices cotranscriptionally, which suggests that TCERG1 forms new macromolecular complexes when splicing occurs cotranscriptionally. In this condition, spliceostatin A has no effect, indicating that TCERG1 rapidly binds and dissociates from stalled spliceosomal complexes and that the mobility properties of TCERG1 do not depend on events occurring after the initial spliceosome formation. Taken together, these data suggest that TCERG1 binds independently to elongation and splicing complexes, thus performing their coupling by transient interactions rather than by stable association with one or the other complexes. This finding has conceptual implications for understanding the coupling between transcription and RNA processing.


Subject(s)
RNA Splicing , Transcription Elongation, Genetic , Transcriptional Elongation Factors/physiology , Cell Nucleus/metabolism , Genes, Reporter , HEK293 Cells , HIV-1/genetics , Humans , Protein Transport
14.
J Virol ; 91(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28122976

ABSTRACT

All retroviruses need to integrate a DNA copy of their genome into the host chromatin. Cellular proteins regulating and targeting lentiviral and gammaretroviral integration in infected cells have been discovered, but the factors that mediate alpharetroviral avian leukosis virus (ALV) integration are unknown. In this study, we have identified the FACT protein complex, which consists of SSRP1 and Spt16, as a principal cellular binding partner of ALV integrase (IN). Biochemical experiments with purified recombinant proteins show that SSRP1 and Spt16 are able to individually bind ALV IN, but only the FACT complex effectively stimulates ALV integration activity in vitro Likewise, in infected cells, the FACT complex promotes ALV integration activity, with proviral integration frequency varying directly with cellular expression levels of the FACT complex. An increase in 2-long-terminal-repeat (2-LTR) circles in the depleted FACT complex cell line indicates that this complex regulates the ALV life cycle at the level of integration. This regulation is shown to be specific to ALV, as disruption of the FACT complex did not inhibit either lentiviral or gammaretroviral integration in infected cells.IMPORTANCE The majority of human gene therapy approaches utilize HIV-1- or murine leukemia virus (MLV)-based vectors, which preferentially integrate near genes and regulatory regions; thus, insertional mutagenesis is a substantial risk. In contrast, ALV integrates more randomly throughout the genome, which decreases the risks of deleterious integration. Understanding how ALV integration is regulated could facilitate the development of ALV-based vectors for use in human gene therapy. Here we show that the FACT complex directly binds and regulates ALV integration efficiency in vitro and in infected cells.


Subject(s)
Avian Leukosis Virus/genetics , Cell Cycle Proteins/physiology , DNA, Viral/physiology , DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Transcription Factors/physiology , Transcriptional Elongation Factors/physiology , Amino Acid Sequence , Animals , Avian Leukosis Virus/enzymology , Chick Embryo , Conserved Sequence , HEK293 Cells , Humans , Integrases/physiology , Protein Binding , Protein Interaction Domains and Motifs , Virus Integration
15.
Mol Cell ; 37(3): 429-37, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20159561

ABSTRACT

Chromosomal translocations involving the MLL gene are associated with infant acute lymphoblastic and mixed lineage leukemia. There are a large number of translocation partners of MLL that share very little sequence or seemingly functional similarities; however, their translocations into MLL result in the pathogenesis of leukemia. To define the molecular reason why these translocations result in the pathogenesis of leukemia, we purified several of the commonly occurring MLL chimeras. We have identified super elongation complex (SEC) associated with all chimeras purified. SEC includes ELL, P-TEFb, AFF4, and several other factors. AFF4 is required for SEC stability and proper transcription by poised RNA polymerase II in metazoans. Knockdown of AFF4 in leukemic cells shows reduction in MLL chimera target gene expression, suggesting that AFF4/SEC could be a key regulator in the pathogenesis of leukemia through many of the MLL partners.


Subject(s)
Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Positive Transcriptional Elongation Factor B/metabolism , Repressor Proteins/physiology , Transcriptional Elongation Factors/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Heat-Shock Response , Histone-Lysine N-Methyltransferase , Homeobox A10 Proteins , Homeodomain Proteins/metabolism , Humans , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein/physiology , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Alignment , Transcription, Genetic , Transcriptional Elongation Factors/physiology , Translocation, Genetic
16.
Mol Cell ; 35(2): 191-205, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19647516

ABSTRACT

Multiple RNA polymerase II (RNAPII) molecules can transcribe a gene simultaneously, but what happens when such polymerases collide--for example due to polymerase pausing or DNA damage? Here, RNAPII collision was characterized using a reconstituted system for simultaneous transcription by two polymerases. When progression of leading polymerase is obstructed, rear-end collision entails a transient state in which the elongation complexes interact, followed by substantial backtracking of trailing polymerase. Elongation complexes remain stable on DNA, with their activity and the integrity of transcription bubbles remaining intact. Subsequent TFIIS-stimulated transcript cleavage allows resumed forward translocation, resulting in trailing polymerase oscillating at the obstruction. Conversely, if leading polymerase is merely stalled at a pause site, collision and TFIIS cooperate to drive it through. We propose that dynamic interactions between RNAPII elongation complexes help regulate polymerase traffic and that their conformational flexibility buffers the effect of collisions with objects on DNA, thereby maintaining stability in the face of obstacles to transcription.


Subject(s)
DNA/metabolism , Models, Genetic , RNA Polymerase II/metabolism , Transcription, Genetic/physiology , Enzyme Stability , Escherichia coli/genetics , RNA Polymerase II/chemistry , Transcriptional Elongation Factors/physiology , Yeasts/genetics
17.
Mol Cell ; 35(3): 377-83, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19683500

ABSTRACT

Nucleosomes are surprisingly dynamic structures in vivo, showing transcription-independent exchange of histones H2A-H2B genome-wide and exchange of H3-H4 mainly within the promoters of transcribed genes. In addition, nucleosomes are disrupted in front of and reassembled behind the elongating RNA polymerase. Here we show that inactivation of histone chaperone Spt16 in yeast results in rapid loss of H2B and H3 from transcribed genes but also from inactive genes. In all cases, histone loss is blocked by a transcription inhibitor, indicating a transcription-dependent event. Thus, nucleosomes are efficiently evicted by the polymerase but do not reform in the absence of Spt16. Yet exchange of nucleosomal H2B with free histones occurs normally, and, unexpectedly, incorporation of new H3 increases at all loci tested. This points to Spt16 restoring normal nucleosome structure by redepositing the displaced H3-H4 histones, thereby preventing incorporation of new histones and perhaps changes in histone modification patterns associated with ongoing transcription.


Subject(s)
DNA-Directed RNA Polymerases/physiology , Histones/metabolism , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Transcriptional Elongation Factors/physiology , Chromatin Assembly and Disassembly/physiology , Histones/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcriptional Elongation Factors/metabolism
18.
Mol Cell ; 35(3): 365-76, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19683499

ABSTRACT

FACT has been proposed to function by displacing H2A-H2B dimers from nucleosomes to form hexasomes. Results described here with yeast FACT (yFACT) suggest instead that nucleosomes are reorganized to a form with the original composition but a looser, more dynamic structure. First, yFACT enhances hydroxyl radical accessibility and endonuclease digestion in vitro at sites throughout the nucleosome, not just in regions contacted by H2A-H2B. Accessibility increases dramatically, but the DNA remains partially protected. Second, increased nuclease sensitivity can occur without displacement of dimers from the nucleosome. Third, yFACT is required for eviction of nucleosomes from the GAL1-10 promoter during transcriptional activation in vivo, but the preferential reduction in dimer occupancy expected for hexasome formation is not observed. We propose that yFACT promotes a reversible transition between two nucleosomal forms, and that this activity contributes to the establishment and maintenance of the chromatin barrier as well as to overcoming it.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Histones/metabolism , Nucleosomes/chemistry , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Transcriptional Elongation Factors/physiology , DNA, Fungal/chemistry , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonucleases, Type II Site-Specific/metabolism , Dimerization , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Models, Genetic , Models, Molecular , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
19.
J Biol Chem ; 290(45): 27297-27310, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26378236

ABSTRACT

Our functional genomic RNAi screens have identified the protein components of the FACT (facilitates chromatin transcription) complex, SUPT16H and SSRP1, as top host factors that negatively regulate HIV-1 replication. FACT interacts specifically with histones H2A/H2B to affect assembly and disassembly of nucleosomes, as well as transcription elongation. We further investigated the suppressive role of FACT proteins in HIV-1 transcription. First, depletion of SUPT16H or SSRP1 protein enhances Tat-mediated HIV-1 LTR (long terminal repeat) promoter activity. Second, HIV-1 Tat interacts with SUPT16H but not SSRP1 protein. However, both SUPT16H and SSRP1 are recruited to LTR promoter. Third, the presence of SUPT16H interferes with the association of Cyclin T1 (CCNT1), a subunit of P-TEFb, with the Tat-LTR axis. Removing inhibitory mechanisms to permit HIV-1 transcription is an initial and key regulatory step to reverse post-integrated latent HIV-1 proviruses for purging of reservoir cells. We therefore evaluated the role of FACT proteins in HIV-1 latency and reactivation. Depletion of SUPT16H or SSRP1 protein affects both HIV-1 transcriptional initiation and elongation and spontaneously reverses latent HIV-1 in U1/HIV and J-LAT cells. Similar effects were observed with a primary CD4+ T cell model of HIV-1 latency. FACT proteins also interfere with HTLV-1 Tax-LTR-mediated transcription and viral latency, indicating that they may act as general transcriptional suppressors for retroviruses. We conclude that FACT proteins SUPT16H and SSRP1 play a key role in suppressing HIV-1 transcription and promoting viral latency, which may serve as promising gene targets for developing novel HIV-1 latency-reversing agents.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , HIV-1/physiology , High Mobility Group Proteins/physiology , Human T-lymphotropic virus 1/physiology , Transcription Factors/physiology , Transcriptional Elongation Factors/physiology , Virus Latency/physiology , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/virology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line , Cyclin T/physiology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , HEK293 Cells , HIV Long Terminal Repeat , HIV-1/genetics , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Human T-lymphotropic virus 1/genetics , Humans , Models, Biological , Positive Transcriptional Elongation Factor B/physiology , Promoter Regions, Genetic , RNA Interference , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Elongation Factors/antagonists & inhibitors , Transcriptional Elongation Factors/genetics , Virus Latency/genetics
20.
J Cell Biochem ; 117(3): 612-20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26264132

ABSTRACT

TCERG1 was characterized previously as a repressor of the transcription factor C/EBPα through a mechanism that involved relocalization of TCERG1 from nuclear speckles to pericentromeric regions. The inhibitory activity as well as the relocalization activity has been demonstrated to lie in the amino terminal half of the protein, which contains several discrete motifs including an imperfect glutamine-alanine (QA) repeat. In the present study, we showed that deletion of this domain completely abrogated the ability of TCERG1 to inhibit the growth arrest activity of C/EBPα. Moreover, the QA repeat deletion mutant of TCERG1 lost the ability to be relocalized from nuclear speckles to pericentromeric regions, and caused an increase in the average size of individual speckles. We also showed that deletion of the QA repeat abrogated the complex formation between TCERG1 and C/EBPα. Examination of mutants with varying numbers of QA repeats indicated that a minimal number of repeats are required for inhibitory activity as well as relocalization ability. These data contribute to our overall understanding of how TCERG1 can have gene-specific effects in addition to its more general roles in coordinating transcription elongation and splicing.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Transcriptional Elongation Factors/chemistry , Animals , COS Cells , Cell Cycle Checkpoints , Cell Proliferation , Chlorocebus aethiops , HEK293 Cells , Humans , Protein Interaction Domains and Motifs , Protein Transport , Transcriptional Elongation Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL