Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.370
Filter
1.
Cell ; 187(6): 1422-1439.e24, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38447573

ABSTRACT

Neutrophils, the most abundant and efficient defenders against pathogens, exert opposing functions across cancer types. However, given their short half-life, it remains challenging to explore how neutrophils adopt specific fates in cancer. Here, we generated and integrated single-cell neutrophil transcriptomes from 17 cancer types (225 samples from 143 patients). Neutrophils exhibited extraordinary complexity, with 10 distinct states including inflammation, angiogenesis, and antigen presentation. Notably, the antigen-presenting program was associated with favorable survival in most cancers and could be evoked by leucine metabolism and subsequent histone H3K27ac modification. These neutrophils could further invoke both (neo)antigen-specific and antigen-independent T cell responses. Neutrophil delivery or a leucine diet fine-tuned the immune balance to enhance anti-PD-1 therapy in various murine cancer models. In summary, these data not only indicate the neutrophil divergence across cancers but also suggest therapeutic opportunities such as antigen-presenting neutrophil delivery.


Subject(s)
Antigen Presentation , Neoplasms , Neutrophils , Animals , Humans , Mice , Antigens, Neoplasm , Leucine/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neutrophils/metabolism , T-Lymphocytes , Single-Cell Gene Expression Analysis
2.
Nat Immunol ; 25(3): 552-561, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38263463

ABSTRACT

The steady flow of lactic acid (LA) from tumor cells to the extracellular space via the monocarboxylate transporter symport system suppresses antitumor T cell immunity. However, LA is a natural energy metabolite that can be oxidized in the mitochondria and could potentially stimulate T cells. Here we show that the lactate-lowering mood stabilizer lithium carbonate (LC) can inhibit LA-mediated CD8+ T cell immunosuppression. Cytoplasmic LA increased the pumping of protons into lysosomes. LC interfered with vacuolar ATPase to block lysosomal acidification and rescue lysosomal diacylglycerol-PKCθ signaling to facilitate monocarboxylate transporter 1 localization to mitochondrial membranes, thus transporting LA into the mitochondria as an energy source for CD8+ T cells. These findings indicate that targeting LA metabolism using LC could support cancer immunotherapy.


Subject(s)
Antimanic Agents , Lactic Acid , Lithium Carbonate , Mitochondria , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Lactic Acid/metabolism , Lithium Carbonate/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/metabolism , Antimanic Agents/pharmacology
3.
Annu Rev Cell Dev Biol ; 38: 263-289, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35609906

ABSTRACT

Covalently closed, single-stranded circular RNAs can be produced from viral RNA genomes as well as from the processing of cellular housekeeping noncoding RNAs and precursor messenger RNAs. Recent transcriptomic studies have surprisingly uncovered that many protein-coding genes can be subjected to backsplicing, leading to widespread expression of a specific type of circular RNAs (circRNAs) in eukaryotic cells. Here, we discuss experimental strategies used to discover and characterize diverse circRNAs at both the genome and individual gene scales. We further highlight the current understanding of how circRNAs are generated and how the mature transcripts function. Some circRNAs act as noncoding RNAs to impact gene regulation by serving as decoys or competitors for microRNAs and proteins. Others form extensive networks of ribonucleoprotein complexes or encode functional peptides that are translated in response to certain cellular stresses. Overall, circRNAs have emerged as an important class of RNAmolecules in gene expression regulation that impact many physiological processes, including early development, immune responses, neurogenesis, and tumorigenesis.


Subject(s)
MicroRNAs , RNA, Circular , Gene Expression Regulation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA/genetics , RNA/metabolism , RNA, Circular/genetics , RNA, Untranslated , RNA, Viral , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism
4.
Cell ; 181(3): 621-636.e22, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259487

ABSTRACT

Long noncoding RNAs (lncRNAs) evolve more rapidly than mRNAs. Whether conserved lncRNAs undergo conserved processing, localization, and function remains unexplored. We report differing subcellular localization of lncRNAs in human and mouse embryonic stem cells (ESCs). A significantly higher fraction of lncRNAs is localized in the cytoplasm of hESCs than in mESCs. This turns out to be important for hESC pluripotency. FAST is a positionally conserved lncRNA but is not conserved in its processing and localization. In hESCs, cytoplasm-localized hFAST binds to the WD40 domain of the E3 ubiquitin ligase ß-TrCP and blocks its interaction with phosphorylated ß-catenin to prevent degradation, leading to activated WNT signaling, required for pluripotency. In contrast, mFast is nuclear retained in mESCs, and its processing is suppressed by the splicing factor PPIE, which is highly expressed in mESCs but not hESCs. These findings reveal that lncRNA processing and localization are previously under-appreciated contributors to the rapid evolution of function.


Subject(s)
Intracellular Space/genetics , RNA, Long Noncoding/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/metabolism , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , RNA Splicing/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Stem Cells/pathology
5.
Cell ; 179(7): 1448-1450, 2019 Dec 12.
Article in English | MEDLINE | ID: mdl-31835025

ABSTRACT

Many targeted base transversions, insertions, and deletions remain challenging due to the lack of precise and efficient genome editing technologies. Recently, Anzalone et al. reported a versatile approach to achieve all types of genome edits, shedding new light on correcting most genetic variants associated with diseases.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Animals , Genetic Therapy/methods , Humans
6.
Cell ; 177(4): 865-880.e21, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31031002

ABSTRACT

Circular RNAs (circRNAs) produced from back-splicing of exons of pre-mRNAs are widely expressed, but current understanding of their functions is limited. These RNAs are stable in general and are thought to have unique structural conformations distinct from their linear RNA cognates. Here, we show that endogenous circRNAs tend to form 16-26 bp imperfect RNA duplexes and act as inhibitors of double-stranded RNA (dsRNA)-activated protein kinase (PKR) related to innate immunity. Upon poly(I:C) stimulation or viral infection, circRNAs are globally degraded by RNase L, a process required for PKR activation in early cellular innate immune responses. Augmented PKR phosphorylation and circRNA reduction are found in peripheral blood mononuclear cells (PBMCs) derived from patients with autoimmune disease systemic lupus erythematosus (SLE). Importantly, overexpression of the dsRNA-containing circRNA in PBMCs or T cells derived from SLE can alleviate the aberrant PKR activation cascade, thus providing a connection between circRNAs and SLE.


Subject(s)
RNA, Circular/metabolism , RNA, Circular/physiology , eIF-2 Kinase/metabolism , Adolescent , Adult , Autoimmune Diseases/genetics , Cell Line , Endoribonucleases/metabolism , Female , Humans , Immunity, Innate/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/genetics , Middle Aged , Phosphorylation , RNA/metabolism , RNA Splicing/genetics , RNA Stability/physiology , RNA, Circular/genetics , RNA, Double-Stranded/metabolism , Virus Diseases/metabolism , eIF-2 Kinase/immunology
7.
Cell ; 169(4): 664-678.e16, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475895

ABSTRACT

Dysregulated rRNA synthesis by RNA polymerase I (Pol I) is associated with uncontrolled cell proliferation. Here, we report a box H/ACA small nucleolar RNA (snoRNA)-ended long noncoding RNA (lncRNA) that enhances pre-rRNA transcription (SLERT). SLERT requires box H/ACA snoRNAs at both ends for its biogenesis and translocation to the nucleolus. Deletion of SLERT impairs pre-rRNA transcription and rRNA production, leading to decreased tumorigenesis. Mechanistically, SLERT interacts with DEAD-box RNA helicase DDX21 via a 143-nt non-snoRNA sequence. Super-resolution images reveal that DDX21 forms ring-shaped structures surrounding multiple Pol I complexes and suppresses pre-rRNA transcription. Binding by SLERT allosterically alters individual DDX21 molecules, loosens the DDX21 ring, and evicts DDX21 suppression on Pol I transcription. Together, our results reveal an important control of ribosome biogenesis by SLERT lncRNA and its regulatory role in DDX21 ring-shaped arrangements acting on Pol I complexes.


Subject(s)
DEAD-box RNA Helicases/metabolism , RNA Polymerase I/metabolism , RNA Precursors/genetics , RNA, Long Noncoding/metabolism , Allosteric Site , Animals , Carcinogenesis , Cell Line , Cell Line, Tumor , DEAD-box RNA Helicases/chemistry , Female , Gene Knockout Techniques , Humans , Mice , Mice, Nude , RNA Precursors/metabolism , Transcription, Genetic
8.
Mol Cell ; 84(12): 2304-2319.e8, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38838666

ABSTRACT

Circular RNAs (circRNAs) are upregulated during neurogenesis. Where and how circRNAs are localized and what roles they play during this process have remained elusive. Comparing the nuclear and cytoplasmic circRNAs between H9 cells and H9-derived forebrain (FB) neurons, we identify that a subset of adenosine (A)-rich circRNAs are restricted in H9 nuclei but exported to cytosols upon differentiation. Such a subcellular relocation of circRNAs is modulated by the poly(A)-binding protein PABPC1. In the H9 nucleus, newly produced (A)-rich circRNAs are bound by PABPC1 and trapped by the nuclear basket protein TPR to prevent their export. Modulating (A)-rich motifs in circRNAs alters their subcellular localization, and introducing (A)-rich circRNAs in H9 cytosols results in mRNA translation suppression. Moreover, decreased nuclear PABPC1 upon neuronal differentiation enables the export of (A)-rich circRNAs, including circRTN4(2,3), which is required for neurite outgrowth. These findings uncover subcellular localization features of circRNAs, linking their processing and function during neurogenesis.


Subject(s)
Active Transport, Cell Nucleus , Adenosine , Cell Nucleus , Neurogenesis , Neurons , Poly(A)-Binding Protein I , RNA, Circular , RNA , RNA, Circular/metabolism , RNA, Circular/genetics , Neurons/metabolism , Adenosine/metabolism , Cell Nucleus/metabolism , Humans , Poly(A)-Binding Protein I/metabolism , Poly(A)-Binding Protein I/genetics , Animals , RNA/metabolism , RNA/genetics , Cell Line , Cell Differentiation , Cytoplasm/metabolism , Prosencephalon/metabolism
9.
Mol Cell ; 83(24): 4445-4460.e7, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37995689

ABSTRACT

The metazoan-specific Integrator complex catalyzes 3' end processing of small nuclear RNAs (snRNAs) and premature termination that attenuates the transcription of many protein-coding genes. Integrator has RNA endonuclease and protein phosphatase activities, but it remains unclear if both are required for complex function. Here, we show IntS6 (Integrator subunit 6) over-expression blocks Integrator function at a subset of Drosophila protein-coding genes, although having no effect on snRNAs or attenuation of other loci. Over-expressed IntS6 titrates protein phosphatase 2A (PP2A) subunits, thereby only affecting gene loci where phosphatase activity is necessary for Integrator function. IntS6 functions analogous to a PP2A regulatory B subunit as over-expression of canonical B subunits, which do not bind Integrator, is also sufficient to inhibit Integrator activity. These results show that the phosphatase module is critical at only a subset of Integrator-regulated genes and point to PP2A recruitment as a tunable step that modulates transcription termination efficiency.


Subject(s)
Drosophila Proteins , Transcription Termination, Genetic , Animals , RNA , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Small Nuclear/genetics , Transcription Factors/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster
10.
Cell ; 163(6): 1400-12, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26607794

ABSTRACT

Extensive cellular heterogeneity exists within specific immune-cell subtypes classified as a single lineage, but its molecular underpinnings are rarely characterized at a genomic scale. Here, we use single-cell RNA-seq to investigate the molecular mechanisms governing heterogeneity and pathogenicity of Th17 cells isolated from the central nervous system (CNS) and lymph nodes (LN) at the peak of autoimmune encephalomyelitis (EAE) or differentiated in vitro under either pathogenic or non-pathogenic polarization conditions. Computational analysis relates a spectrum of cellular states in vivo to in-vitro-differentiated Th17 cells and unveils genes governing pathogenicity and disease susceptibility. Using knockout mice, we validate four new genes: Gpr65, Plzp, Toso, and Cd5l (in a companion paper). Cellular heterogeneity thus informs Th17 function in autoimmunity and can identify targets for selective suppression of pathogenic Th17 cells while potentially sparing non-pathogenic tissue-protective ones.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Sequence Analysis, RNA , Single-Cell Analysis , Th17 Cells/metabolism , Th17 Cells/pathology , Animals , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Profiling , Humans , Kruppel-Like Transcription Factors/metabolism , Lymph Nodes/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/metabolism , Peptide Fragments/metabolism , Promyelocytic Leukemia Zinc Finger Protein , Receptors, G-Protein-Coupled/metabolism , Receptors, Immunologic/metabolism , Receptors, Scavenger , Th17 Cells/immunology
11.
Mol Cell ; 82(2): 420-434.e6, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34951963

ABSTRACT

Exon back-splicing-generated circular RNAs, as a group, can suppress double-stranded RNA (dsRNA)-activated protein kinase R (PKR) in cells. We have sought to synthesize immunogenicity-free, short dsRNA-containing RNA circles as PKR inhibitors. Here, we report that RNA circles synthesized by permuted self-splicing thymidylate synthase (td) introns from T4 bacteriophage or by Anabaena pre-tRNA group I intron could induce an immune response. Autocatalytic splicing introduces ∼74 nt td or ∼186 nt Anabaena extraneous fragments that can distort the folding status of original circular RNAs or form structures themselves to provoke innate immune responses. In contrast, synthesized RNA circles produced by T4 RNA ligase without extraneous fragments exhibit minimized immunogenicity. Importantly, directly ligated circular RNAs that form short dsRNA regions efficiently suppress PKR activation 103- to 106-fold higher than reported chemical compounds C16 and 2-AP, highlighting the future use of circular RNAs as potent inhibitors for diseases related to PKR overreaction.


Subject(s)
Protein Kinase Inhibitors/pharmacology , RNA, Circular/pharmacology , eIF-2 Kinase/antagonists & inhibitors , A549 Cells , Bacteriophage T4/enzymology , Bacteriophage T4/genetics , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/drug effects , Introns , Nucleic Acid Conformation , Protein Kinase Inhibitors/immunology , RNA Ligase (ATP)/genetics , RNA Ligase (ATP)/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Circular/genetics , RNA, Circular/immunology , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , eIF-2 Kinase/metabolism
12.
Cell ; 159(7): 1488-9, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525868

ABSTRACT

Microexons are frequently underestimated in transcriptome analyses. Two studies published in Cell and Genome Research now independently report the identification of hundreds of microexons. Alternative splicing of some microexons is regulated by neuronal-specific RNA-binding proteins and modifies the function of proteins involved in neurogenesis, with misregulation linked to autism.

13.
Cell ; 159(1): 134-147, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25242744

ABSTRACT

Exon circularization has been identified from many loci in mammals, but the detailed mechanism of its biogenesis has remained elusive. By using genome-wide approaches and circular RNA recapitulation, we demonstrate that exon circularization is dependent on flanking intronic complementary sequences. Such sequences and their distribution exhibit rapid evolutionary changes, showing that exon circularization is evolutionarily dynamic. Strikingly, exon circularization efficiency can be regulated by competition between RNA pairing across flanking introns or within individual introns. Importantly, alternative formation of inverted repeated Alu pairs and the competition between them can lead to alternative circularization, resulting in multiple circular RNA transcripts produced from a single gene. Collectively, exon circularization mediated by complementary sequences in human introns and the potential to generate alternative circularization products extend the complexity of mammalian posttranscriptional regulation.


Subject(s)
Alternative Splicing , Exons , Genome, Human , Alu Elements , Animals , Base Sequence , Embryonic Stem Cells/metabolism , Evolution, Molecular , Humans , Introns , Mammals/genetics , Molecular Sequence Data , Nucleic Acid Conformation , Sequence Alignment
14.
Nature ; 615(7952): 526-534, 2023 03.
Article in English | MEDLINE | ID: mdl-36890225

ABSTRACT

The nucleolus is the most prominent membraneless condensate in the nucleus. It comprises hundreds of proteins with distinct roles in the rapid transcription of ribosomal RNA (rRNA) and efficient processing within units comprising a fibrillar centre and a dense fibrillar component and ribosome assembly in a granular component1. The precise localization of most nucleolar proteins and whether their specific localization contributes to the radial flux of pre-rRNA processing have remained unknown owing to insufficient resolution in imaging studies2-5. Therefore, how these nucleolar proteins are functionally coordinated with stepwise pre-rRNA processing requires further investigation. Here we screened 200 candidate nucleolar proteins using high-resolution live-cell microscopy and identified 12 proteins that are enriched towards the periphery of the dense fibrillar component (PDFC). Among these proteins, unhealthy ribosome biogenesis 1 (URB1) is a static, nucleolar protein that ensures 3' end pre-rRNA anchoring and folding for U8 small nucleolar RNA recognition and the subsequent removal of the 3' external transcribed spacer (ETS) at the dense fibrillar component-PDFC boundary. URB1 depletion leads to a disrupted PDFC, uncontrolled pre-rRNA movement, altered pre-rRNA conformation and retention of the 3' ETS. These aberrant 3' ETS-attached pre-rRNA intermediates activate exosome-dependent nucleolar surveillance, resulting in decreased 28S rRNA production, head malformations in zebrafish and delayed embryonic development in mice. This study provides insight into functional sub-nucleolar organization and identifies a physiologically essential step in rRNA maturation that requires the static protein URB1 in the phase-separated nucleolus.


Subject(s)
Cell Nucleolus , Exosomes , RNA Precursors , RNA Processing, Post-Transcriptional , RNA, Ribosomal , Zebrafish , Animals , Mice , Cell Nucleolus/metabolism , Embryonic Development , Exosomes/metabolism , Head/abnormalities , Microscopy , Nuclear Proteins/metabolism , RNA Precursors/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Ribosomal, 28S/metabolism , Zebrafish/genetics , Zebrafish/metabolism
15.
Nat Methods ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965442

ABSTRACT

Dynamic imaging of genomic loci is key for understanding gene regulation, but methods for imaging genomes, in particular non-repetitive DNAs, are limited. We developed CRISPRdelight, a DNA-labeling system based on endonuclease-deficient CRISPR-Cas12a (dCas12a), with an engineered CRISPR array to track DNA location and motion. CRISPRdelight enables robust imaging of all examined 12 non-repetitive genomic loci in different cell lines. We revealed the confined movement of the CCAT1 locus (chr8q24) at the nuclear periphery for repressed expression and active motion in the interior nucleus for transcription. We uncovered the selective repositioning of HSP gene loci to nuclear speckles, including a remarkable relocation of HSPH1 (chr13q12) for elevated transcription during stresses. Combining CRISPR-dCas12a and RNA aptamers allowed multiplex imaging of four types of satellite DNA loci with a single array, revealing their spatial proximity to the nucleolus-associated domain. CRISPRdelight is a user-friendly and robust system for imaging and tracking genomic dynamics and regulation.

16.
PLoS Biol ; 22(6): e3002680, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38865309

ABSTRACT

CRISPR-Cas12a, often regarded as a precise genome editor, still requires improvements in specificity. In this study, we used a GFP-activation assay to screen 14 new Cas12a nucleases for mammalian genome editing, successfully identifying 9 active ones. Notably, these Cas12a nucleases prefer pyrimidine-rich PAMs. Among these nucleases, we extensively characterized Mb4Cas12a obtained from Moraxella bovis CCUG 2133, which recognizes a YYN PAM (Y = C or T). Our biochemical analysis demonstrates that Mb4Cas12a can cleave double-strand DNA across a wide temperature range. To improve specificity, we constructed a SWISS-MODEL of Mb4Cas12a based on the FnCas12a crystal structure and identified 8 amino acids potentially forming hydrogen bonds at the target DNA-crRNA interface. By replacing these amino acids with alanine to disrupt the hydrogen bond, we tested the influence of each mutation on Mb4Cas12a specificity. Interestingly, the F370A mutation improved specificity with minimal influence on activity. Further study showed that Mb4Cas12a-F370A is capable of discriminating single-nucleotide polymorphisms. These new Cas12a orthologs and high-fidelity variants hold substantial promise for therapeutic applications.


Subject(s)
Alleles , CRISPR-Associated Proteins , CRISPR-Cas Systems , Gene Editing , Gene Editing/methods , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , Humans , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/chemistry , Animals , Protein Engineering/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Polymorphism, Single Nucleotide , Mutation , DNA/metabolism , DNA/genetics , HEK293 Cells
17.
Mol Cell ; 76(5): 767-783.e11, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31540874

ABSTRACT

Fibrillar centers (FCs) and dense fibrillar components (DFCs) are essential morphologically distinct sub-regions of mammalian cell nucleoli for rDNA transcription and pre-rRNA processing. Here, we report that a human nucleolus consists of several dozen FC/DFC units, each containing 2-3 transcriptionally active rDNAs at the FC/DFC border. Pre-rRNA processing factors, such as fibrillarin (FBL), form 18-24 clusters that further assemble into the DFC surrounding the FC. Mechanistically, the 5' end of nascent 47S pre-rRNA binds co-transcriptionally to the RNA-binding domain of FBL. FBL diffuses to the DFC, where local self-association via its glycine- and arginine-rich (GAR) domain forms phase-separated clusters to immobilize FBL-interacting pre-rRNA, thus promoting directional traffic of nascent pre-rRNA while facilitating pre-rRNA processing and DFC formation. These results unveil FC/DFC ultrastructures in nucleoli and suggest a conceptual framework for considering nascent RNA sorting using multivalent interactions of their binding proteins.


Subject(s)
Cell Nucleolus/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , Active Transport, Cell Nucleus , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Cell Nucleolus/genetics , Cell Nucleolus/ultrastructure , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Nucleic Acid Conformation , Protein Binding , Protein Interaction Domains and Motifs , RNA Precursors/genetics , RNA Precursors/ultrastructure , RNA, Ribosomal/genetics , RNA, Ribosomal/ultrastructure
18.
Proc Natl Acad Sci U S A ; 121(7): e2322375121, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38315835

ABSTRACT

Protein S-acyl transferases (PATs) catalyze S-acylation, a reversible post-translational modification critical for membrane association, trafficking, and stability of substrate proteins. Many plant proteins are potentially S-acylated but few have corresponding PATs identified. By using genomic editing, confocal imaging, pharmacological, genetic, and biochemical assays, we demonstrate that three Arabidopsis class C PATs positively regulate BR signaling through S-acylation of BRASSINOSTEROID-SIGNALING KINASE1 (BSK1). PAT19, PAT20, and PAT22 associate with the plasma membrane (PM) and the trans-Golgi network/early endosome (TGN/EE). Functional loss of all three genes results in a plethora of defects, indicative of reduced BR signaling and rescued by enhanced BR signaling. PAT19, PAT20, and PAT22 interact with BSK1 and are critical for the S-acylation of BSK1, and for BR signaling. The PM abundance of BSK1 was reduced by functional loss of PAT19, PAT20, and PAT22 whereas abolished by its S-acylation-deficient point mutations, suggesting a key role of S-acylation in its PM targeting. Finally, an active BR analog induces vacuolar trafficking and degradation of PAT19, PAT20, or PAT22, suggesting that the S-acylation of BSK1 by the three PATs serves as a negative feedback module in BR signaling.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Protein Serine-Threonine Kinases , Acylation , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Brassinosteroids/metabolism , Gene Expression Regulation, Plant , Signal Transduction , Transferases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
19.
Trends Genet ; 39(12): 897-907, 2023 12.
Article in English | MEDLINE | ID: mdl-37839990

ABSTRACT

Numerous circular RNAs (circRNAs) produced from back-splicing of exon(s) have been recently revealed on a genome-wide scale across species. Although generally expressed at a low level, some relatively abundant circRNAs can play regulatory roles in various biological processes, prompting continuous profiling of circRNA in broader conditions. Over the past decade, distinct strategies have been applied in both transcriptome enrichment and bioinformatic tools for detecting and quantifying circRNAs. Understanding the scope and limitations of these strategies is crucial for the subsequent annotation and characterization of circRNAs, especially those with functional potential. Here, we provide an overview of different transcriptome enrichment, deep sequencing and computational approaches for genome-wide circRNA identification, and discuss strategies for accurate quantification and characterization of circRNA.


Subject(s)
RNA, Circular , RNA , RNA, Circular/genetics , RNA/genetics , Transcriptome , Computational Biology , Genome/genetics
20.
Development ; 150(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-37073949

ABSTRACT

De novo root regeneration (DNRR) is a developmental process that regenerates adventitious roots from wounded tissues. Phytohormone signaling pathways involved in microbial resistance are mobilized after cutting and influence de novo root regeneration. Microbes may positively or negatively influence the development and stress responses of a plant. However, most studies on the molecular mechanisms of de novo organogenesis are performed in aseptic conditions. Thus, the potential crosstalk between organ regeneration and biotic stresses is underexplored. Here, we report the development of a versatile experimental system to study the impact of microbes on DNRR. Using this system, we found that bacteria inhibited root regeneration by activation of, but not limited to, pathogen-associated molecular pattern (PAMP)-triggered immunity. Sensing bacteria-derived flagellin 22 peptide (flg22) inhibited root regeneration by interfering with the formation of an auxin maximum at the wound site. This inhibition relies on the receptor complex that recognizes microbial patterns but may bypass the requirement of salicylic acid signaling.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Plant Growth Regulators/metabolism , Indoleacetic Acids/metabolism , Plant Roots/metabolism , Gene Expression Regulation, Plant
SELECTION OF CITATIONS
SEARCH DETAIL