Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Plant Physiol ; 195(2): 1333-1346, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38446745

ABSTRACT

Transposable elements (TEs) contribute to plant evolution, development, and adaptation to environmental changes, but the regulatory mechanisms are largely unknown. RNA-directed DNA methylation (RdDM) is 1 TE regulatory mechanism in plants. Here, we identified that novel ARGONAUTE 1 (AGO1)-binding Tudor domain proteins Precocious dissociation of sisters C/E (PDS5C/E) are involved in 24-nt siRNA production to establish RdDM on TEs in Arabidopsis thaliana. PDS5 family proteins are subunits of the eukaryote-conserved cohesin complex. However, the double mutant lacking angiosperm-specific subfamily PDS5C and PDS5E (pds5c/e) exhibited different developmental phenotypes and transcriptome compared with those of the double mutant lacking eukaryote-conserved subfamily PDS5A and PDS5B (pds5a/b), suggesting that the angiosperm-specific PDS5C/E subfamily has a unique function in angiosperm plants. Proteome and imaging analyses revealed that PDS5C/E interact with AGO1. The pds5c/e double mutant had defects in 24-nt siRNA accumulation and CHH DNA methylation on TEs. In addition, some lncRNAs that accumulated in the pds5c/e mutant were targeted by AGO1-loading 21-nt miRNAs and 21-nt siRNAs. These results indicate that PDS5C/E and AGO1 participate in 24-nt siRNA production for RdDM in the cytoplasm. These findings indicate that angiosperm plants evolved a new regulator, the PDS5C/E subfamily, to control the increase in TEs during angiosperm evolution.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Argonaute Proteins , DNA Methylation , RNA, Small Interfering , DNA Methylation/genetics , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Gene Expression Regulation, Plant , Tudor Domain/genetics , DNA Transposable Elements/genetics , RNA, Plant/genetics , RNA, Plant/metabolism , Mutation/genetics
2.
J Mol Biol ; 436(7): 168413, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38135180

ABSTRACT

KAT8 is an evolutionarily conserved lysine acetyltransferase that catalyzes histone acetylation at H4K16 or H4K5 and H4K8 through distinct protein complexes. It plays a pivotal role in male X chromosome dosage compensation in Drosophila and is implicated in the regulation of diverse cellular processes in mammals. Mutations and dysregulation of KAT8 have been reported in human neurodevelopmental disorders and various cancers. However, the precise mechanisms by which these mutations disrupt KAT8's normal function, leading to disease pathogenesis, remain largely unknown. In this study, we focus on a hotspot missense cancer mutation, the R98W point mutation within the Tudor-knot domain. Our study reveals that the R98W mutation leads to a reduction in global H4K16ac levels in cells and downregulates the expression of target genes. Mechanistically, we demonstrate that R98 is essential for KAT8-mediated acetylation of nucleosomal histones by modulating substrate accessibility.


Subject(s)
Histone Acetyltransferases , Histones , Neoplasms , Nucleosomes , Tudor Domain , Animals , Humans , Male , Acetylation , Drosophila/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/genetics , Histones/metabolism , Neoplasms/genetics , Mutation, Missense , Nucleosomes/metabolism , Tudor Domain/genetics , Cell Line, Tumor
3.
Life Sci Alliance ; 4(3)2021 03.
Article in English | MEDLINE | ID: mdl-33376130

ABSTRACT

Aub guided by piRNAs ensures genome integrity by cleaving retrotransposons, and genome propagation by trapping mRNAs to form the germplasm that instructs germ cell formation. Arginines at the N-terminus of Aub (Aub-NTRs) interact with Tudor and other Tudor domain-containing proteins (TDRDs). Aub-TDRD interactions suppress active retrotransposons via piRNA amplification and form germplasm via generation of Aub-Tudor ribonucleoproteins. Here, we show that Aub-NTRs are dispensable for primary piRNA biogenesis but essential for piRNA amplification and that their symmetric dimethylation is required for germplasm formation and germ cell specification but largely redundant for piRNA amplification.


Subject(s)
Argonaute Proteins/metabolism , Cytoplasm/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Amplification , Germ Cells/metabolism , Peptide Initiation Factors/metabolism , RNA, Small Interfering/genetics , Tudor Domain/genetics , Animals , Animals, Genetically Modified , Arginine/metabolism , DNA Transposable Elements/genetics , Drosophila Proteins/genetics , Female , Male , Ovary/metabolism , Peptide Initiation Factors/genetics , RNA, Messenger/metabolism
4.
Commun Biol ; 3(1): 607, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097771

ABSTRACT

All living organisms have to cope with the constant threat of genome damage by UV light and other toxic reagents. To maintain the integrity of their genomes, organisms developed a variety of DNA repair pathways. One of these, the Transcription Coupled DNA-Repair (TCR) pathway, is triggered by stalled RNA Polymerase (RNAP) complexes at DNA damage sites on actively transcribed genes. A recently elucidated bacterial TCR pathway employs the UvrD helicase pulling back stalled RNAP complexes from the damage, stimulating recruitment of the DNA-repair machinery. However, structural and functional aspects of UvrD's interaction with RNA Polymerase remain elusive. Here we used advanced solution NMR spectroscopy to investigate UvrD's role within the TCR, identifying that the carboxy-terminal region of the UvrD helicase facilitates RNAP interactions by adopting a Tudor-domain like fold. Subsequently, we functionally analyzed this domain, identifying it as a crucial component for the UvrD-RNAP interaction besides having nucleic-acid affinity.


Subject(s)
DNA Helicases , DNA Repair/genetics , DNA-Directed RNA Polymerases , Escherichia coli Proteins , Tudor Domain/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA Damage/genetics , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Protein Folding , Staphylococcus aureus/genetics
5.
EMBO J ; 39(20): e105130, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32914505

ABSTRACT

Silkworm ovarian germ cells produce the Siwi-piRNA-induced silencing complex (piRISC) through two consecutive mechanisms, the primary pathway and the secondary ping-pong cycle. Primary Siwi-piRISC production occurs on the outer mitochondrial membrane in an Ago3-independent manner, where Tudor domain-containing Papi binds unloaded Siwi via its symmetrical dimethylarginines (sDMAs). Here, we now show that secondary Siwi-piRISC production occurs at the Ago3-positive nuage Ago3 bodies, in an Ago3-dependent manner, where Vreteno (Vret), another Tudor protein, interconnects unloaded Siwi and Ago3-piRISC through their sDMAs. Upon Siwi depletion, Ago3 is phosphorylated and insolubilized in its piRISC form with cleaved RNAs and Vret, suggesting that the complex is stalled in the intermediate state. The Ago3 bodies are also enlarged. The aberrant morphology is restored upon Siwi re-expression without Ago3-piRISC supply. Thus, Siwi depletion aggregates the Ago3 bodies to protect the piRNA intermediates from degradation until the normal cellular environment returns to re-initiate the ping-pong cycle. Overall, these findings reveal a unique regulatory mechanism controlling piRNA biogenesis.


Subject(s)
Argonaute Proteins/metabolism , Bombyx/metabolism , Germ Cells/metabolism , Insect Proteins/metabolism , RNA, Small Interfering/metabolism , Tudor Domain/genetics , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Argonaute Proteins/genetics , Bombyx/genetics , Bombyx/growth & development , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chromatography, Liquid , Computational Biology , Female , Insect Proteins/genetics , Ovary/cytology , Ovary/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , RNA-Seq , Tandem Mass Spectrometry
6.
J Mol Biol ; 432(14): 4061-4075, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32428527

ABSTRACT

Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is an essential factor for the maintenance of mammalian DNA methylation and harbors several reader modules for recognizing epigenetic marks. The tandem Tudor domain (TTD) of UHRF1 has a peptide-binding groove that functions as a binding platform for intra- or intermolecular interactions. Besides the groove interacting with unphosphorylated linker 2 and spacer of UHRF1, it also interacts with di/tri-methylated histone H3 at Lys9 and DNA ligase 1 (LIG1) at Lys126. Here we focus on the phosphorylation of Ser298 in linker 2, which was implied to regulate the ligand-binding property of the TTD. Although the protein expression level of UHRF1 is unchanged throughout the cell cycle, Ser298 phosphorylated form of UHRF1 is notably increased in the G2/M phase, which is revealed by immunoprecipitation followed by Western blotting. Molecularly, while unphosphorylated linker 2 covers the peptide-binding groove to prevent access of other interactors, small-angle X-ray scattering, thermal stability assay and molecular dynamics simulation revealed that the phosphate group of Ser298 dissociates linker 2 from the peptide-binding groove of the TTD to permit the other interactors to access to the groove. Our data reveal a mechanism in which Ser298 phosphorylation in linker 2 triggers a change of the TTD's structure and may affect multiple functions of UHRF1 by facilitating associations with LIG1 at DNA replication sites and histone H3K9me2/me3 at heterochromatic regions.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , DNA Methylation/genetics , DNA Replication/genetics , Tudor Domain/genetics , Ubiquitin-Protein Ligases/genetics , DNA, Intergenic/genetics , Epigenesis, Genetic/genetics , Histones/genetics , Humans , Ligands , Molecular Dynamics Simulation , Phosphorylation/genetics , Protein Binding/genetics , Scattering, Small Angle , Serine/genetics
7.
Elife ; 92020 04 27.
Article in English | MEDLINE | ID: mdl-32338603

ABSTRACT

piRNAs play a critical role in the regulation of transposons and other germline genes. In Caenorhabditis elegans, regulation of piRNA target genes is mediated by the mutator complex, which synthesizes high levels of siRNAs through the activity of an RNA-dependent RNA polymerase. However, the steps between mRNA recognition by the piRNA pathway and siRNA amplification by the mutator complex are unknown. Here, we identify the Tudor domain protein, SIMR-1, as acting downstream of piRNA production and upstream of mutator complex-dependent siRNA biogenesis. Interestingly, SIMR-1 also localizes to distinct subcellular foci adjacent to P granules and Mutator foci, two phase-separated condensates that are the sites of piRNA-dependent mRNA recognition and mutator complex-dependent siRNA amplification, respectively. Thus, our data suggests a role for multiple perinuclear condensates in organizing the piRNA pathway and promoting mRNA regulation by the mutator complex.


In the biological world, a process known as RNA interference helps cells to switch genes on and off and to defend themselves against harmful genetic material. This mechanism works by deactivating RNA sequences, the molecular templates cells can use to create proteins. Overall, RNA interference relies on the cell creating small RNA molecules that can target and inhibit the harmful RNA sequences that need to be silenced. More precisely, in round worms such as Caenorhabditis elegans, RNA interference happens in two steps. First, primary small RNAs identify the target sequences, which are then combatted by newly synthetised, secondary small RNAs. A number of proteins are also involved in both steps of the process. RNA interference is particularly important to preserve fertility, guarding sex cells against 'rogue' segments of genetic information that could be passed on to the next generation. In future sex cells, the proteins involved in RNA interference cluster together, forming a structure called a germ granule. Yet, little is known about the roles and identity of these proteins. To fill this knowledge gap, Manage et al. focused on the second stage of the RNA interference pathway in the germ granules of C. elegans, examining the molecules that physically interact with a key protein. This work revealed a new protein called SIMR-1. Looking into the role of SIMR-1 showed that the protein is required to amplify secondary small RNAs, but not to identify target sequences. However, it only promotes the creation of secondary small RNAs if a specific subtype of primary small RNAs have recognized the target RNAs for silencing. Further experiments also showed that within the germ granule, SIMR-1 is present in a separate substructure different from any compartment previously identified. This suggests that each substep of the RNA interference process takes place at a different location in the granule. In both C. elegans and humans, disruptions in the RNA interference pathway can lead to conditions such as cancer or infertility. Dissecting the roles of the proteins involved in this process in roundworms may help to better grasp how this process unfolds in mammals, and how it could be corrected in the case of disease.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Tudor Domain/genetics , Animals , Female , Male
8.
J Struct Biol ; 209(1): 107406, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31747559

ABSTRACT

The essential SAS2-related acetyltransferase 1 (Esa1), as a acetyltransferase of MYST family, is indispensable for the cell cycle and transcriptional regulation. The Tudor domain consists of 60 amino acids and belongs to the Royal family, which serves as a module interacting with methylated histone and/or DNA. Although Tudor domain has been widely studied in higher eukaryotes, its structure and function remain unclear in Trypanosoma brucei (T. brucei), a protozoan unicellular parasite causing sleeping sickness in human and nagana in cattle in sub-Saharan Africa. Here, we determined a high-resolution structure of TbEsa1 presumed Tudor domain from T. brucei by X-ray crystallography. TbEsa1 Tudor domain adopts a conserved Tudor-like fold, which is comprised of a five-stranded ß-barrel surrounded by two short α-helices. Furthermore, we revealed a non-specific DNA binding pattern of TbEsa1 Tudor domain. However, TbEsa1 Tudor domain showed no methyl-histone binding ability, due to the absence of key aromatic residues forming a conserved aromatic cage.


Subject(s)
Histone Acetyltransferases/ultrastructure , Trypanosoma brucei brucei/ultrastructure , Trypanosomiasis, African/microbiology , Tudor Domain/genetics , Amino Acid Sequence/genetics , Animals , Binding Sites/genetics , Cattle , Crystallography, X-Ray , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Humans , Models, Molecular , Protein Binding/genetics , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/genetics , Trypanosomiasis, African/enzymology , Trypanosomiasis, African/genetics
9.
Nucleic Acids Res ; 47(5): 2546-2559, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30566680

ABSTRACT

PIWI-interacting RNAs (piRNAs) comprise a class of small RNAs best known for suppressing transposable elements in germline tissues. The vector mosquito Aedes aegypti encodes seven PIWI genes, four of which are somatically expressed. This somatic piRNA pathway generates piRNAs from viral RNA during infection with cytoplasmic RNA viruses through ping-pong amplification by the PIWI proteins Ago3 and Piwi5. Yet, additional insights into the molecular mechanisms mediating non-canonical piRNA production are lacking. TUDOR-domain containing (Tudor) proteins facilitate piRNA biogenesis in Drosophila melanogaster and other model organisms. We thus hypothesized that Tudor proteins are required for viral piRNA production and performed a knockdown screen targeting all A. aegypti Tudor genes. Knockdown of the Tudor genes AAEL012437, Vreteno, Yb, SMN and AAEL008101-RB resulted in significantly reduced viral piRNA levels, with AAEL012437-depletion having the strongest effect. This protein, which we named Veneno, associates directly with Ago3 in an sDMA-dependent manner and localizes in cytoplasmic foci reminiscent of piRNA processing granules of Drosophila. Veneno-interactome analyses reveal a network of co-factors including the orthologs of the Drosophila piRNA pathway components Vasa and Yb, which in turn interacts with Piwi5. We propose that Veneno assembles a multi-protein complex for ping-pong dependent piRNA production from viral RNA.


Subject(s)
Aedes/genetics , Drosophila Proteins/genetics , RNA, Small Interfering/genetics , Tudor Domain/genetics , Aedes/pathogenicity , Animals , Argonaute Proteins/genetics , DNA Transposable Elements/genetics , Drosophila melanogaster/genetics , Germ Cells/growth & development , Mosquito Vectors/genetics , Multiprotein Complexes/genetics
10.
Nat Commun ; 9(1): 2123, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844495

ABSTRACT

P53-binding protein 1 (53BP1) regulates the double-strand break (DSB) repair pathway choice. A recently identified 53BP1-binding protein Tudor-interacting repair regulator (TIRR) modulates the access of 53BP1 to DSBs by masking the H4K20me2 binding surface on 53BP1, but the underlying mechanism remains unclear. Here we report the 1.76-Å crystal structure of TIRR in complex with 53BP1 tandem Tudor domain. We demonstrate that the N-terminal region (residues 10-24) and the L8-loop of TIRR interact with 53BP1 Tudor through three loops (L1, L3, and L1'). TIRR recognition blocks H4K20me2 binding to 53BP1 Tudor and modulates 53BP1 functions in vivo. Structure comparisons identify a TIRR histidine (H106) that is absent from the TIRR homolog Nudt16, but essential for 53BP1 Tudor binding. Remarkably, mutations mimicking TIRR binding modules restore the disrupted binding of Nudt16-53BP1 Tudor. Our studies elucidate the mechanism by which TIRR recognizes 53BP1 Tudor and functions as a cellular inhibitor of the histone methyl-lysine readers.


Subject(s)
Carrier Proteins/metabolism , Protein Structure, Tertiary/physiology , Tudor Domain/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Animals , Binding Sites/genetics , Cell Line, Tumor , Crystallography, X-Ray , DNA Breaks, Double-Stranded , DNA Repair/physiology , HEK293 Cells , Humans , Mice , Protein Binding/genetics , RNA-Binding Proteins , Tumor Suppressor p53-Binding Protein 1/genetics
11.
Proc Natl Acad Sci U S A ; 113(38): E5544-51, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27582462

ABSTRACT

Topoisomerase 3ß (Top3ß) can associate with the mediator protein Tudor domain-containing protein 3 (TDRD3) to participate in two gene expression processes of transcription and translation. Despite the apparent importance of TDRD3 in binding with Top3ß and directing it to cellular compartments critical for gene expression, the biochemical mechanism of how TDRD3 can affect the functions of Top3ß is not known. We report here sensitive biochemical assays for the activities of Top3ß on DNA and RNA substrates in resolving topological entanglements and for the analysis of TDRD3 functions. TDRD3 stimulates the relaxation activity of Top3ß on hypernegatively supercoiled DNA and changes the reaction from a distributive to a processive mode. Both supercoil retention assays and binding measurement by fluorescence anisotropy reveal a heretofore unknown preference for binding single-stranded nucleic acids over duplex. Whereas TDRD3 has a structure-specific binding preference, it does not discriminate between DNA and RNA. This unique property for binding with nucleic acids can have an important function in serving as a hub to form nucleoprotein complexes on DNA and RNA. To gain insight into the roles of Top3ß on RNA metabolism, we designed an assay by annealing two single-stranded RNA circles with complementary sequences. Top3ß is capable of converting two such single-stranded RNA circles into a double-stranded RNA circle, and this strand-annealing activity is enhanced by TDRD3. These results demonstrate that TDRD3 can enhance the biochemical activities of Top3ß on both DNA and RNA substrates, in addition to its function of targeting Top3ß to critical sites in subcellular compartments.


Subject(s)
DNA Topoisomerases/genetics , DNA, Superhelical/genetics , Drosophila Proteins/genetics , Nucleoproteins/genetics , Amino Acid Sequence/genetics , Animals , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , DNA, Superhelical/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Drosophila/genetics , Gene Expression Regulation/genetics , Macromolecular Substances/chemistry , Nucleoproteins/chemistry , Protein Binding , Protein Biosynthesis , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , Transcription, Genetic , Tudor Domain/genetics
12.
Nucleic Acids Res ; 44(13): 6102-12, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27016734

ABSTRACT

Histone post-translational modifications, and specific combinations they create, mediate a wide range of nuclear events. However, the mechanistic bases for recognition of these combinations have not been elucidated. Here, we characterize crosstalk between H3T3 and H3T6 phosphorylation, occurring in mitosis, and H3K4me3, a mark associated with active transcription. We detail the molecular mechanisms by which H3T3ph/K4me3/T6ph switches mediate activities of H3K4me3-binding proteins, including those containing plant homeodomain (PHD) and double Tudor reader domains. Our results derived from nuclear magnetic resonance chemical shift perturbation analysis, orthogonal binding assays and cell fluorescence microscopy studies reveal a strong anti-correlation between histone H3T3/T6 phosphorylation and retention of PHD finger proteins in chromatin during mitosis. Together, our findings uncover the mechanistic rules of chromatin engagement for H3K4me3-specific readers during cell division.


Subject(s)
Chromatin/genetics , Heterochromatin/genetics , Mitosis/genetics , Protein Processing, Post-Translational/genetics , Histone Code/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Methylation , Phosphorylation , Protein Binding/genetics , Tudor Domain/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...