Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 348
Filter
1.
Mol Cell ; 84(7): 1191-1205.e7, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38458202

ABSTRACT

Polycomb repressive complex 1 (PRC1) is a key transcriptional regulator in development via modulating chromatin structure and catalyzing histone H2A ubiquitination at Lys119 (H2AK119ub1). H2AK119ub1 is one of the most abundant histone modifications in mammalian cells. However, the function of H2AK119ub1 in polycomb-mediated gene silencing remains debated. In this study, we reveal that H2AK119ub1 has two distinct roles in gene expression, through differentially modulating chromatin compaction mediated by canonical PRC1 and the linker histone H1. Interestingly, we find that H2AK119ub1 plays a positive role in transcription through interfering with the binding of canonical PRC1 to nucleosomes and therefore counteracting chromatin condensation. Conversely, we demonstrate that H2AK119ub1 facilitates H1-dependent chromatin condensation and enhances the silencing of developmental genes in mouse embryonic stem cells, suggesting that H1 may be one of several possible pathways for H2AK119ub1 in repressing transcription. These results provide insights and molecular mechanisms by which H2AK119ub1 differentially fine-tunes developmental gene expression.


Subject(s)
Chromatin , Polycomb Repressive Complex 1 , Animals , Mice , Chromatin/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Nucleosomes/genetics , Ubiquitination , Gene Expression , Mammals/metabolism
2.
Mol Cell ; 83(16): 2884-2895.e7, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37536340

ABSTRACT

DNA replication ensures the accurate transmission of genetic information during the cell cycle. Histone variant H2A.Z is crucial for early replication origins licensing and activation in which SUV420H1 preferentially recognizes H2A.Z-nucleosome and deposits H4 lysine 20 dimethylation (H4K20me2) on replication origins. Here, we report the cryo-EM structures of SUV420H1 bound to H2A.Z-nucleosome or H2A-nucleosome and demonstrate that SUV420H1 directly interacts with H4 N-terminal tail, the DNA, and the acidic patch in the nucleosome. The H4 (1-24) forms a lasso-shaped structure that stabilizes the SUV420H1-nucleosome complex and precisely projects the H4K20 residue into the SUV420H1 catalytic center. In vitro and in vivo analyses reveal a crucial role of the SUV420H1 KR loop (residues 214-223), which lies close to the H2A.Z-specific residues D97/S98, in H2A.Z-nucleosome preferential recognition. Together, our findings elucidate how SUV420H1 recognizes nucleosomes to ensure site-specific H4K20me2 modification and provide insights into how SUV420H1 preferentially recognizes H2A.Z nucleosome.


Subject(s)
Histones , Nucleosomes , Histones/metabolism , Nucleosomes/genetics , Methylation , DNA/metabolism , DNA Replication
3.
Mol Cell ; 82(6): 1169-1185.e7, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35202573

ABSTRACT

Polycomb group (PcG) proteins are essential for post-implantation development by depositing repressive histone modifications at promoters, mainly CpG islands (CGIs), of developmental regulator genes. However, promoter PcG marks are erased after fertilization and de novo established in peri-implantation embryos, coinciding with the transition from naive to primed pluripotency. Nevertheless, the molecular basis for this establishment remains unknown. In this study, we show that the expression of the long KDM2B isoform (KDM2BLF), which contains the demethylase domain, is specifically induced at peri-implantation and that its H3K36me2 demethylase activity is required for PcG enrichment at CGIs. Moreover, KDM2BLF interacts with BRG1/BRM-associated factor (BAF) and stabilizes BAF occupancy at CGIs for subsequent gain of accessibility, which precedes PcG enrichment. Consistently, KDM2BLF inactivation results in significantly delayed post-implantation development. In summary, our data unveil dynamic chromatin configuration of CGIs during exit from naive pluripotency and provide a conceptual framework for the spatiotemporal establishment of PcG functions.


Subject(s)
Chromatin , Drosophila Proteins , CpG Islands , Drosophila Proteins/metabolism , Histone Code , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic
4.
Mol Cell ; 82(21): 4018-4032.e9, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36332605

ABSTRACT

Kinetochore assembly on centromeres is central for chromosome segregation, and defects in this process cause mitotic errors and aneuploidy. Besides the well-established protein network, emerging evidence suggests the involvement of regulatory RNA in kinetochore assembly; however, it has remained elusive about the identity of such RNA, let alone its mechanism of action in this critical process. Here, we report CCTT, a previously uncharacterized long non-coding RNA (lncRNA) transcribed from the arm of human chromosome 17, which plays a vital role in kinetochore assembly. We show that CCTT highly localizes to all centromeres via the formation of RNA-DNA triplex and specifically interacts with CENP-C to help engage this blueprint protein in centromeres, and consequently, CCTT loss triggers extensive mitotic errors and aneuploidy. These findings uncover a non-centromere-derived lncRNA that recruits CENP-C to centromeres and shed critical lights on the function of centromeric DNA sequences as anchor points for kinetochore assembly.


Subject(s)
RNA, Long Noncoding , Humans , Aneuploidy , Centromere Protein A/metabolism , DNA , Kinetochores/metabolism , RNA, Long Noncoding/genetics , Centromere
5.
Nature ; 616(7955): 176-182, 2023 04.
Article in English | MEDLINE | ID: mdl-36991118

ABSTRACT

Repression of gene expression by protein complexes of the Polycomb group is a fundamental mechanism that governs embryonic development and cell-type specification1-3. The Polycomb repressive deubiquitinase (PR-DUB) complex removes the ubiquitin moiety from monoubiquitinated histone H2A K119 (H2AK119ub1) on the nucleosome4, counteracting the ubiquitin E3 ligase activity of Polycomb repressive complex 1 (PRC1)5 to facilitate the correct silencing of genes by Polycomb proteins and safeguard active genes from inadvertent silencing by PRC1 (refs. 6-9). The intricate biological function of PR-DUB requires accurate targeting of H2AK119ub1, but PR-DUB can deubiquitinate monoubiquitinated free histones and peptide substrates indiscriminately; the basis for its exquisite nucleosome-dependent substrate specificity therefore remains unclear. Here we report the cryo-electron microscopy structure of human PR-DUB, composed of BAP1 and ASXL1, in complex with the chromatosome. We find that ASXL1 directs the binding of the positively charged C-terminal extension of BAP1 to nucleosomal DNA and histones H3-H4 near the dyad, an addition to its role in forming the ubiquitin-binding cleft. Furthermore, a conserved loop segment of the catalytic domain of BAP1 is situated near the H2A-H2B acidic patch. This distinct nucleosome-binding mode displaces the C-terminal tail of H2A from the nucleosome surface, and endows PR-DUB with the specificity for H2AK119ub1.


Subject(s)
Deubiquitinating Enzymes , Histones , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Humans , Cryoelectron Microscopy , Histones/chemistry , Histones/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/ultrastructure , Polycomb-Group Proteins/chemistry , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/ultrastructure , Ubiquitin/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/ultrastructure , Ubiquitination , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Repressor Proteins/ultrastructure , Catalytic Domain , Deubiquitinating Enzymes/classification , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/ultrastructure , Substrate Specificity , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/ultrastructure
6.
Genes Dev ; 35(23-24): 1610-1624, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34819355

ABSTRACT

Chromosomal duplication requires de novo assembly of nucleosomes from newly synthesized histones, and the process involves a dynamic network of interactions between histones and histone chaperones. sNASP and ASF1 are two major histone H3-H4 chaperones found in distinct and common complexes, yet how sNASP binds H3-H4 in the presence and absence of ASF1 remains unclear. Here we show that, in the presence of ASF1, sNASP principally recognizes a partially unfolded Nα region of histone H3, and in the absence of ASF1, an additional sNASP binding site becomes available in the core domain of the H3-H4 complex. Our study also implicates a critical role of the C-terminal tail of H4 in the transfer of H3-H4 between sNASP and ASF1 and the coiled-coil domain of sNASP in nucleosome assembly. These findings provide mechanistic insights into coordinated histone binding and transfer by histone chaperones.


Subject(s)
Histone Chaperones , Histones , Binding Sites , Cell Cycle Proteins/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Nucleosomes , Protein Binding
7.
Cell ; 152(4): 859-72, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23415232

ABSTRACT

Histone modifications are key regulators of chromatin function. However, little is known to what extent histone modifications can directly impact on chromatin. Here, we address how a modification within the globular domain of histones regulates chromatin function. We demonstrate that H3K122ac can be sufficient to stimulate transcription and that mutation of H3K122 impairs transcriptional activation, which we attribute to a direct effect of H3K122ac on histone-DNA binding. In line with this, we find that H3K122ac defines genome-wide genetic elements and chromatin features associated with active transcription. Furthermore, H3K122ac is catalyzed by the coactivators p300/CBP and can be induced by nuclear hormone receptor signaling. Collectively, this suggests that transcriptional regulators elicit their effects not only via signaling to histone tails but also via direct structural perturbation of nucleosomes by directing acetylation to their lateral surface.


Subject(s)
Gene Expression Regulation , Histone Code , Histones/metabolism , Transcriptional Activation , Acetylation , Animals , Cell Line, Tumor , Eukaryota/metabolism , Fibroblasts/metabolism , Humans , Mice , Models, Molecular , Nucleosomes/metabolism , Receptors, Estrogen/metabolism , Schizosaccharomyces/metabolism , Transcription Initiation Site , p300-CBP Transcription Factors/metabolism
8.
Cell ; 153(5): 1012-24, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706739

ABSTRACT

Histone acetylation plays critical roles in chromatin remodeling, DNA repair, and epigenetic regulation of gene expression, but the underlying mechanisms are unclear. Proteasomes usually catalyze ATP- and polyubiquitin-dependent proteolysis. Here, we show that the proteasomes containing the activator PA200 catalyze the polyubiquitin-independent degradation of histones. Most proteasomes in mammalian testes ("spermatoproteasomes") contain a spermatid/sperm-specific α subunit α4 s/PSMA8 and/or the catalytic ß subunits of immunoproteasomes in addition to PA200. Deletion of PA200 in mice abolishes acetylation-dependent degradation of somatic core histones during DNA double-strand breaks and delays core histone disappearance in elongated spermatids. Purified PA200 greatly promotes ATP-independent proteasomal degradation of the acetylated core histones, but not polyubiquitinated proteins. Furthermore, acetylation on histones is required for their binding to the bromodomain-like regions in PA200 and its yeast ortholog, Blm10. Thus, PA200/Blm10 specifically targets the core histones for acetylation-mediated degradation by proteasomes, providing mechanisms by which acetylation regulates histone degradation, DNA repair, and spermatogenesis.


Subject(s)
DNA Repair , Histones/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Spermatogenesis , Testis/metabolism , Acetylation , Amino Acid Sequence , Animals , DNA Breaks, Double-Stranded , Humans , Male , Mice , Molecular Sequence Data , Nuclear Proteins/chemistry , Protein Structure, Tertiary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment
9.
Nature ; 611(7936): 614-622, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36323776

ABSTRACT

Sperm chromatin is typically transformed by protamines into a compact and transcriptionally inactive state1,2. Sperm cells of flowering plants lack protamines, yet they have small, transcriptionally active nuclei with chromatin condensed through an unknown mechanism3,4. Here we show that a histone variant, H2B.8, mediates sperm chromatin and nuclear condensation in Arabidopsis thaliana. Loss of H2B.8 causes enlarged sperm nuclei with dispersed chromatin, whereas ectopic expression in somatic cells produces smaller nuclei with aggregated chromatin. This result demonstrates that H2B.8 is sufficient for chromatin condensation. H2B.8 aggregates transcriptionally inactive AT-rich chromatin into phase-separated condensates, which facilitates nuclear compaction without reducing transcription. Reciprocal crosses show that mutation of h2b.8 reduces male transmission, which suggests that H2B.8-mediated sperm compaction is important for fertility. Altogether, our results reveal a new mechanism of nuclear compaction through global aggregation of unexpressed chromatin. We propose that H2B.8 is an evolutionary innovation of flowering plants that achieves nuclear condensation compatible with active transcription.


Subject(s)
Arabidopsis , Cell Size , Chromatin , Histones , Pollen , Arabidopsis/cytology , Arabidopsis/genetics , Arabidopsis/metabolism , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Histones/classification , Histones/genetics , Histones/metabolism , Protamines , Pollen/cytology , Pollen/genetics , Pollen/metabolism , Gene Expression Regulation, Plant , AT Rich Sequence , Cell Nucleus/genetics , Mutation , Cell Nucleus Size , Phase Transition , Transcription, Genetic
10.
Plant Cell ; 36(5): 1829-1843, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38309957

ABSTRACT

In the eukaryotic nucleus, heterochromatin forms highly condensed, visible foci known as heterochromatin foci (HF). These HF are enriched with linker histone H1, a key player in heterochromatin condensation and silencing. However, it is unknown how H1 aggregates HF and condenses heterochromatin. In this study, we established that H1 facilitates heterochromatin condensation by enhancing inter- and intrachromosomal interactions between and within heterochromatic regions of the Arabidopsis (Arabidopsis thaliana) genome. We demonstrated that H1 drives HF formation via phase separation, which requires its C-terminal intrinsically disordered region (C-IDR). A truncated H1 lacking the C-IDR fails to form foci or recover HF in the h1 mutant background, whereas C-IDR with a short stretch of the globular domain (18 out of 71 amino acids) is sufficient to rescue both defects. In addition, C-IDR is essential for H1's roles in regulating nucleosome repeat length and DNA methylation in Arabidopsis, indicating that phase separation capability is required for chromatin functions of H1. Our data suggest that bacterial H1-like proteins, which have been shown to condense DNA, are intrinsically disordered and capable of mediating phase separation. Therefore, we propose that phase separation mediated by H1 or H1-like proteins may represent an ancient mechanism for condensing chromatin and DNA.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Heterochromatin , Histones , Arabidopsis/genetics , Arabidopsis/metabolism , Heterochromatin/metabolism , Heterochromatin/genetics , Histones/metabolism , Histones/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , DNA Methylation/genetics , Nucleosomes/metabolism , Phase Separation
11.
Mol Cell ; 75(1): 102-116.e9, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31128943

ABSTRACT

Transcription regulation underlies stem cell function and development. Here, we elucidate an unexpected role of an essential ribogenesis factor, WDR43, as a chromatin-associated RNA-binding protein (RBP) and release factor in modulating the polymerase (Pol) II activity for pluripotency regulation. WDR43 binds prominently to promoter-associated noncoding/nascent RNAs, occupies thousands of gene promoters and enhancers, and interacts with the Pol II machinery in embryonic stem cells (ESCs). Nascent transcripts and transcription recruit WDR43 to active promoters, where WDR43 facilitates releases of the elongation factor P-TEFb and paused Pol II. Knockdown of WDR43 causes genome-wide defects in Pol II release and pluripotency-associated gene expression. Importantly, auxin-mediated rapid degradation of WDR43 drastically reduces Pol II activity, precluding indirect consequences. These results reveal an RNA-mediated recruitment and feedforward regulation on transcription and demonstrate an unforeseen role of an RBP in promoting Pol II elongation and coordinating high-level transcription and translation in ESC pluripotency.


Subject(s)
Cation Transport Proteins/genetics , Chromatin/chemistry , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , RNA Polymerase II/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Transcription, Genetic , Zebrafish Proteins/genetics , Animals , Binding Sites , Cation Transport Proteins/metabolism , Cell Differentiation , Cell Line , Chromatin/metabolism , Embryo, Mammalian , Enhancer Elements, Genetic , Gene Deletion , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Biosynthesis , Proteolysis , RNA Polymerase II/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Zebrafish Proteins/metabolism
12.
Mol Cell ; 76(4): 646-659.e6, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31543422

ABSTRACT

Eukaryotic chromosomes contain compartments of various functions, which are marked by and enriched with specific histone modifications. However, the molecular mechanisms by which these histone marks function in chromosome compartmentalization are poorly understood. Constitutive heterochromatin is a largely silent chromosome compartment characterized in part by H3K9me2 and 3. Here, we show that heterochromatin protein 1 (HP1), an H3K9me2 and 3 "reader," interacts with SUV39H1, an H3K9me2 and 3 "writer," and with TRIM28, an abundant HP1 scaffolding protein, to form complexes with increased multivalent engagement of H3K9me2 and 3-modified chromatin. H3K9me2 and 3-marked nucleosomal arrays and associated complexes undergo phase separation to form macromolecule-enriched liquid droplets. The droplets are reminiscent of heterochromatin as they are highly dense chromatin-containing structures that are resistant to DNase and exclude the general transcription factor TFIIB. Our data suggest a general mechanism by which histone marks regulate chromosome compartmentalization by promoting phase separation.


Subject(s)
Chromatin Assembly and Disassembly , Heterochromatin/metabolism , Histones/metabolism , Lipid Droplets/metabolism , Nucleosomes/metabolism , Protein Processing, Post-Translational , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , HEK293 Cells , Heterochromatin/genetics , Humans , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Multiprotein Complexes , Nucleosomes/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Time Factors , Tripartite Motif-Containing Protein 28/genetics , Tripartite Motif-Containing Protein 28/metabolism
13.
Nature ; 578(7793): E8, 2020 02.
Article in English | MEDLINE | ID: mdl-31932733

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
Nature ; 577(7791): 576-581, 2020 01.
Article in English | MEDLINE | ID: mdl-31875854

ABSTRACT

DNA replication is a tightly regulated process that ensures the precise duplication of the genome during the cell cycle1. In eukaryotes, the licensing and activation of replication origins are regulated by both DNA sequence and chromatin features2. However, the chromatin-based regulatory mechanisms remain largely uncharacterized. Here we show that, in HeLa cells, nucleosomes containing the histone variant H2A.Z are enriched with histone H4 that is dimethylated on its lysine 20 residue (H4K20me2) and with bound origin-recognition complex (ORC). In vitro studies show that H2A.Z-containing nucleosomes bind directly to the histone lysine methyltransferase enzyme SUV420H1, promoting H4K20me2 deposition, which is in turn required for ORC1 binding. Genome-wide studies show that signals from H4K20me2, ORC1 and nascent DNA strands co-localize with H2A.Z, and that depletion of H2A.Z results in decreased H4K20me2, ORC1 and nascent-strand signals throughout the genome. H2A.Z-regulated replication origins have a higher firing efficiency and early replication timing compared with other origins. Our results suggest that the histone variant H2A.Z epigenetically regulates the licensing and activation of early replication origins and maintains replication timing through the SUV420H1-H4K20me2-ORC1 axis.


Subject(s)
DNA Replication Timing , DNA Replication , Histones/metabolism , Replication Origin/genetics , DNA/metabolism , DNA Replication/genetics , Epigenesis, Genetic , HeLa Cells , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Humans , Lysine/metabolism , Methylation , Nucleosomes/chemistry , Nucleosomes/metabolism , Origin Recognition Complex/metabolism
15.
Mol Cell ; 71(2): 284-293.e4, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30029006

ABSTRACT

The human FACT (facilitates chromatin transcription) complex, composed of two subunits SPT16 (Suppressor of Ty 16) and SSRP1 (Structure-specific recognition protein-1), plays essential roles in nucleosome remodeling. However, the molecular mechanism of FACT reorganizing the nucleosome still remains elusive. In this study, we demonstrate that FACT displays dual functions in destabilizing the nucleosome and maintaining the original histones and nucleosome integrity at the single-nucleosome level. We found that the subunit SSRP1 is responsible for maintenance of nucleosome integrity by holding the H3/H4 tetramer on DNA and promoting the deposition of the H2A/H2B dimer onto the nucleosome. In contrast, the large subunit SPT16 destabilizes the nucleosome structure by displacing the H2A/H2B dimers. Our findings provide mechanistic insights by which the two subunits of FACT coordinate with each other to fulfill its functions and suggest that FACT may play essential roles in preserving the original histones with epigenetic identity during transcription or DNA replication.


Subject(s)
DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Nucleosomes/metabolism , Transcriptional Elongation Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/metabolism , DNA/metabolism , DNA Replication , DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Histones/metabolism , Humans , Models, Molecular , Nucleosomes/genetics , Protein Binding , Protein Multimerization , Saccharomyces cerevisiae Proteins/metabolism , Sf9 Cells , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/genetics
16.
Chem Soc Rev ; 53(4): 1892-1914, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38230701

ABSTRACT

Molecular assembly is the process of organizing individual molecules into larger structures and complex systems. The self-assembly approach is predominantly utilized in creating artificial molecular assemblies, and was believed to be the primary mode of molecular assembly in living organisms as well. However, it has been shown that the assembly of many biological complexes is "catalysed" by other molecules, rather than relying solely on self-assembly. In this review, we summarize these catalysed-assembly (catassembly) phenomena in living organisms and systematically analyse their mechanisms. We then expand on these phenomena and discuss related concepts, including catalysed-disassembly and catalysed-reassembly. Catassembly proves to be an efficient and highly selective strategy for synergistically controlling and manipulating various noncovalent interactions, especially in hierarchical molecular assemblies. Overreliance on self-assembly may, to some extent, hinder the advancement of artificial molecular assembly with powerful features. Furthermore, inspired by the biological catassembly phenomena, we propose guidelines for designing artificial catassembly systems and developing characterization and theoretical methods, and review pioneering works along this new direction. Overall, this approach may broaden and deepen our understanding of molecular assembly, enabling the construction and control of intelligent assembly systems with advanced functionality.

17.
Mol Phylogenet Evol ; 197: 108082, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38705251

ABSTRACT

In addition to topography and climate, biogeographic dispersal has been considered to influence plant diversity in the Himalaya-Hengduan Mountains (HHM), yet, the mode and tempo of sky island dispersal and its influence on species richness has been little explored. Through phylogenetic analysis of Gaultheria ser. Trichophyllae, a sky island alpine clade within the HHM, we test the hypothesis that dispersal has affected current local species richness. We inferred the dynamics of biogeographic dispersal with correlation tests on direction, distance, occurrence time, and regional species richness. We found that G. ser. Trichophyllae originated at the end of the Miocene and mostly dispersed toward higher longitudes (eastward). In particular, shorter intra-regional eastward dispersals and longer inter-regional westward dispersals were most frequently observed. We detected a prevalence of eastward intra-region dispersals in both glacial periods and interglacials. These dispersals may have been facilitated by the reorganization of paleo-drainages and monsoon intensification through time. We suggest that the timing of dispersal corresponding to glacial periods and the prevalence of intra-region dispersal, rather than dispersal frequency, most influenced the pattern of species richness of G. ser. Trichophyllae. This study facilitates a more comprehensive understanding of biodiversity in the sky islands within the HHM.


Subject(s)
Biodiversity , Phylogeny , China , Phylogeography , Islands , Plant Dispersal
18.
Microb Pathog ; 193: 106768, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960217

ABSTRACT

Fowl cholera is an infectious disease that affects both poultry and wild birds, characterized by hemorrhagic and septicemic symptoms, caused by Pasteurella multocida (P. multocida), and leading to substantial economic losses in the poultry sector. The development of genetic engineering vaccines against avian P. multocida encountered early-stage challenges due to the limited availability of effective gene editing tools. Presently, NgAgoDM-enhanced homologous recombination stands as a potent technique for achieving efficient gene knockout in avian P. multocida. Hence, this study employed NgAgoDM-enhanced homologous recombination to target and knockout hyaE (239-359aa), hyaD, hexABC, and hexD, denoted as ΔhyaE (239-359aa), ΔhyaD, ΔhexABC, and ΔhexD, respectively. Additionally, we generated a hyaD recovery strain with two point mutations, designated as mhyaD. Thus, this study systematically examined the impact of capsular synthetic gene clusters on the pathogenicity of P. multocida. Moreover, the study demonstrated the critical role of hyaD activity in the virulence of avian P. multocida. This study offers novel insights for enhancing attenuated vaccines further.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Poultry Diseases , Pasteurella multocida/genetics , Pasteurella multocida/pathogenicity , Animals , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Virulence/genetics , Poultry Diseases/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Homologous Recombination , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Gene Knockout Techniques , Chickens/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Birds/microbiology , Multigene Family , Virulence Factors/genetics , Poultry/microbiology
19.
Nat Chem Biol ; 18(1): 70-80, 2022 01.
Article in English | MEDLINE | ID: mdl-34916619

ABSTRACT

An RNA-involved phase-separation model has been proposed for transcription control. However, the molecular links that connect RNA to the transcription machinery remain missing. Here we find that RNA-binding proteins (RBPs) constitute half of the chromatin proteome in embryonic stem cells (ESCs), some being colocalized with RNA polymerase (Pol) II at promoters and enhancers. Biochemical analyses of representative RBPs show that the paraspeckle protein PSPC1 inhibits the RNA-induced premature release of Pol II, and makes use of RNA as multivalent molecules to enhance the formation of transcription condensates and subsequent phosphorylation and release of Pol II. This synergistic interplay enhances polymerase engagement and activity via the RNA-binding and phase-separation activities of PSPC1. In ESCs, auxin-induced acute degradation of PSPC1 leads to genome-wide defects in Pol II binding and nascent transcription. We propose that promoter-associated RNAs and their binding proteins synergize the phase separation of polymerase condensates to promote active transcription.


Subject(s)
RNA Polymerase II/metabolism , RNA-Binding Proteins/metabolism , Transcription, Genetic , Gene Expression Regulation , Phosphorylation , Promoter Regions, Genetic , Protein Binding
20.
Phytopathology ; 114(3): 500-502, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37750871

ABSTRACT

The strains in Lysobacter spp. have the potential to control plant-parasitic nematodes. In our experiment, L. gummosus YMF3.00690 showed antagonistic effects against plant root-knot nematode. Nine metabolites were isolated and identified from cultures of L. gummosus YMF3.00690, of which compound 1 was identified as a new metabolite tetrahydro-4,4,6-trimethyl-6-[(tetrahydro-6,6-dimethyl-2-oxo-4(1H)-pyrimidinylidene) methyl]-2(1H)-pyrimidinone. The activity assay showed that two compounds, 5-(hydroxymethyl)-1H-pyrrole-2-carbaldehyde (2) and 1H-pyrrole-2-carboxylic acid (3), had nematicidal activities against Meloidogyne javanica with mortalities of 69.93 and 90.54% at 400 ppm for 96 h, respectively. These two compounds were further tested for the inhibition activity of eggs hatching, and compound 3 showed a significant inhibition rate of 63.36% at 50 ppm for 48 h. In the chemotactic activity assay, three compounds (1 to 3) were found to have concentration-dependent chemotactic activity, of which compound 1 showed attractive activity. This experiment explored the active metabolites of L. gummosus YMF3.00690 against M. javanica and laid the foundation for biopesticide development.


Subject(s)
Lysobacter , Tylenchoidea , Animals , Tylenchoidea/physiology , Plant Diseases/prevention & control , Plant Diseases/parasitology , Antinematodal Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL