Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.772
Filter
Add more filters

Publication year range
1.
Cell ; 187(13): 3262-3283.e23, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38815580

ABSTRACT

In eukaryotes, the Suv39 family of proteins tri-methylate lysine 9 of histone H3 (H3K9me) to form constitutive heterochromatin. However, how Suv39 proteins are nucleated at heterochromatin is not fully described. In the fission yeast, current models posit that Argonaute1-associated small RNAs (sRNAs) nucleate the sole H3K9 methyltransferase, Clr4/SUV39H, to centromeres. Here, we show that in the absence of all sRNAs and H3K9me, the Mtl1 and Red1 core (MTREC)/PAXT complex nucleates Clr4/SUV39H at a heterochromatic long noncoding RNA (lncRNA) at which the two H3K9 deacetylases, Sir2 and Clr3, also accumulate by distinct mechanisms. Iterative cycles of H3K9 deacetylation and methylation spread Clr4/SUV39H from the nucleation center in an sRNA-independent manner, generating a basal H3K9me state. This is acted upon by the RNAi machinery to augment and amplify the Clr4/H3K9me signal at centromeres to establish heterochromatin. Overall, our data reveal that lncRNAs and RNA quality control factors can nucleate heterochromatin and function as epigenetic silencers in eukaryotes.


Subject(s)
Cell Cycle Proteins , Heterochromatin , Histone-Lysine N-Methyltransferase , Histones , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Cell Cycle Proteins/metabolism , Centromere/metabolism , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methylation , Methyltransferases/metabolism , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/metabolism , RNA, Fungal/genetics , RNA, Small Interfering/genetics
2.
Cell ; 185(12): 2132-2147.e26, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35688134

ABSTRACT

RNA quality control relies on co-factors and adaptors to identify and prepare substrates for degradation by ribonucleases such as the 3' to 5' ribonucleolytic RNA exosome. Here, we determined cryogenic electron microscopy structures of human nuclear exosome targeting (NEXT) complexes bound to RNA that reveal mechanistic insights to substrate recognition and early steps that precede RNA handover to the exosome. The structures illuminate ZCCHC8 as a scaffold, mediating homodimerization while embracing the MTR4 helicase and flexibly anchoring RBM7 to the helicase core. All three subunits collaborate to bind the RNA, with RBM7 and ZCCHC8 surveying sequences upstream of the 3' end to facilitate RNA capture by MTR4. ZCCHC8 obscures MTR4 surfaces important for RNA binding and extrusion as well as MPP6-dependent recruitment and docking onto the RNA exosome core, interactions that contribute to RNA surveillance by coordinating RNA capture, translocation, and extrusion from the helicase to the exosome for decay.


Subject(s)
Exosomes , DEAD-box RNA Helicases/metabolism , DNA Helicases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosomes/metabolism , Humans , Nuclear Proteins/metabolism , Protein Binding , RNA/metabolism , RNA Stability
3.
Annu Rev Cell Dev Biol ; 36: 237-264, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32749865

ABSTRACT

Parkinson's disease (PD) is a leading cause of neurodegeneration that is defined by the selective loss of dopaminergic neurons and the accumulation of protein aggregates called Lewy bodies (LBs). The unequivocal identification of Mendelian inherited mutations in 13 genes in PD has provided transforming insights into the pathogenesis of this disease. The mechanistic analysis of several PD genes, including α-synuclein (α-syn), leucine-rich repeat kinase 2 (LRRK2), PTEN-induced kinase 1 (PINK1), and Parkin, has revealed central roles for protein aggregation, mitochondrial damage, and defects in endolysosomal trafficking in PD neurodegeneration. In this review, we outline recent advances in our understanding of these gene pathways with a focus on the emergent role of Rab (Ras analog in brain) GTPases and vesicular trafficking as a common mechanism that underpins how mutations in PD genes lead to neuronal loss. These advances have led to previously distinct genes such as vacuolar protein-sorting-associated protein 35 (VPS35) and LRRK2 being implicated in a common signaling pathway. A greater understanding of these common nodes of vesicular trafficking will be crucial for linking other PD genes and improving patient stratification in clinical trials underway against α-syn and LRRK2 targets.


Subject(s)
Parkinson Disease/metabolism , Animals , Autophagy , Cytoplasmic Vesicles/metabolism , Humans , Mitochondria/metabolism , Parkinson Disease/genetics , Protein Aggregates , Protein Transport
4.
Cell ; 175(3): 695-708.e13, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30293865

ABSTRACT

We have uncovered the existence of extracellular vesicle (EV)-mediated signaling between cell types within the adipose tissue (AT) proper. This phenomenon became evident in our attempts at generating an adipocyte-specific knockout of caveolin 1 (cav1) protein. Although we effectively ablated the CAV1 gene in adipocytes, cav1 protein remained abundant. With the use of newly generated mouse models, we show that neighboring endothelial cells (ECs) transfer cav1-containing EVs to adipocytes in vivo, which reciprocate by releasing EVs to ECs. AT-derived EVs contain proteins and lipids capable of modulating cellular signaling pathways. Furthermore, this mechanism facilitates transfer of plasma constituents from ECs to the adipocyte. The transfer event is physiologically regulated by fasting/refeeding and obesity, suggesting EVs participate in the tissue response to changes in the systemic nutrient state. This work offers new insights into the complex signaling mechanisms that exist among adipocytes, stromal vascular cells, and, potentially, distal organs.


Subject(s)
Adipocytes/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Fasting/metabolism , Signal Transduction , Animals , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line , Cells, Cultured , Endothelium, Vascular/cytology , Male , Mice , Mice, Inbred C57BL
5.
Cell ; 172(1-2): 275-288.e18, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29328916

ABSTRACT

The neuronal gene Arc is essential for long-lasting information storage in the mammalian brain, mediates various forms of synaptic plasticity, and has been implicated in neurodevelopmental disorders. However, little is known about Arc's molecular function and evolutionary origins. Here, we show that Arc self-assembles into virus-like capsids that encapsulate RNA. Endogenous Arc protein is released from neurons in extracellular vesicles that mediate the transfer of Arc mRNA into new target cells, where it can undergo activity-dependent translation. Purified Arc capsids are endocytosed and are able to transfer Arc mRNA into the cytoplasm of neurons. These results show that Arc exhibits similar molecular properties to retroviral Gag proteins. Evolutionary analysis indicates that Arc is derived from a vertebrate lineage of Ty3/gypsy retrotransposons, which are also ancestors to retroviruses. These findings suggest that Gag retroelements have been repurposed during evolution to mediate intercellular communication in the nervous system.


Subject(s)
Cytoskeletal Proteins/metabolism , Exosomes/metabolism , Gene Products, gag/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Animals , Cells, Cultured , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Endocytosis , Female , Gene Products, gag/chemistry , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/physiology
6.
Cell ; 173(7): 1663-1677.e21, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29906447

ABSTRACT

The ribonucleolytic RNA exosome interacts with RNA helicases to degrade RNA. To understand how the 3' to 5' Mtr4 helicase engages RNA and the nuclear exosome, we reconstituted 14-subunit Mtr4-containing RNA exosomes from Saccharomyces cerevisiae, Schizosaccharomyces pombe, and human and show that they unwind structured substrates to promote degradation. We loaded a human exosome with an optimized DNA-RNA chimera that stalls MTR4 during unwinding and determined its structure to an overall resolution of 3.45 Å by cryoelectron microscopy (cryo-EM). The structure reveals an RNA-engaged helicase atop the non-catalytic core, with RNA captured within the central channel and DIS3 exoribonuclease active site. MPP6 tethers MTR4 to the exosome through contacts to the RecA domains of MTR4. EXOSC10 remains bound to the core, but its catalytic module and cofactor C1D are displaced by RNA-engaged MTR4. Competition for the exosome core may ensure that RNA is committed to degradation by DIS3 when engaged by MTR4.


Subject(s)
DNA Helicases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , RNA Helicases/metabolism , RNA/metabolism , Catalytic Domain , Cryoelectron Microscopy , DNA/genetics , DNA/metabolism , Exoribonucleases/chemistry , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/chemistry , Humans , Image Processing, Computer-Assisted , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Binding , Protein Structure, Quaternary , RNA/genetics , RNA Helicases/chemistry , RNA Stability , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Substrate Specificity
7.
Cell ; 169(3): 523-537.e15, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431250

ABSTRACT

The distribution of sense and antisense strand DNA mutations on transcribed duplex DNA contributes to the development of immune and neural systems along with the progression of cancer. Because developmentally matured B cells undergo biologically programmed strand-specific DNA mutagenesis at focal DNA/RNA hybrid structures, they make a convenient system to investigate strand-specific mutagenesis mechanisms. We demonstrate that the sense and antisense strand DNA mutagenesis at the immunoglobulin heavy chain locus and some other regions of the B cell genome depends upon localized RNA processing protein complex formation in the nucleus. Both the physical proximity and coupled activities of RNA helicase Mtr4 (and senataxin) with the noncoding RNA processing function of RNA exosome determine the strand-specific distribution of DNA mutations. Our study suggests that strand-specific DNA mutagenesis-associated mechanisms will play major roles in other undiscovered aspects of organismic development.


Subject(s)
B-Lymphocytes/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Mutation , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Nucleus/metabolism , DNA Helicases/metabolism , Exoribonucleases/genetics , Genomic Instability , Immunoglobulin Heavy Chains/genetics , Mice , Multifunctional Enzymes , Nuclear Proteins/genetics , RNA Helicases , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics
8.
Cell ; 169(4): 679-692.e14, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475896

ABSTRACT

The nuclear RNA exosome is an essential multi-subunit complex that controls RNA homeostasis. Congenital mutations in RNA exosome genes are associated with neurodegenerative diseases. Little is known about the role of the RNA exosome in the cellular response to pathogens. Here, using NGS and human and mouse genetics, we show that influenza A virus (IAV) ribogenesis and growth are suppressed by impaired RNA exosome activity. Mechanistically, the nuclear RNA exosome coordinates the initial steps of viral transcription with RNAPII at host promoters. The viral polymerase complex co-opts the nuclear RNA exosome complex and cellular RNAs en route to 3' end degradation. Exosome deficiency uncouples chromatin targeting of the viral polymerase complex and the formation of cellular:viral RNA hybrids, which are essential RNA intermediates that license transcription of antisense genomic viral RNAs. Our results suggest that evolutionary arms races have shaped the cellular RNA quality control machinery.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/virology , RNA Polymerase II/metabolism , A549 Cells , Animals , Chromatin Immunoprecipitation , Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Exosomes/metabolism , Humans , Mass Spectrometry , Mice , Mutation , Neurodegenerative Diseases/virology , RNA-Binding Proteins/genetics , Ribosomes/genetics , Transcription, Genetic
9.
Mol Cell ; 84(11): 2070-2086.e20, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38703770

ABSTRACT

The MYCN oncoprotein binds active promoters in a heterodimer with its partner protein MAX. MYCN also interacts with the nuclear exosome, a 3'-5' exoribonuclease complex, suggesting a function in RNA metabolism. Here, we show that MYCN forms stable high-molecular-weight complexes with the exosome and multiple RNA-binding proteins. MYCN binds RNA in vitro and in cells via a conserved sequence termed MYCBoxI. In cells, MYCN associates with thousands of intronic transcripts together with the ZCCHC8 subunit of the nuclear exosome targeting complex and enhances their processing. Perturbing exosome function results in global re-localization of MYCN from promoters to intronic RNAs. On chromatin, MYCN is then replaced by the MNT(MXD6) repressor protein, inhibiting MYCN-dependent transcription. RNA-binding-deficient alleles show that RNA-binding limits MYCN's ability to activate cell growth-related genes but is required for MYCN's ability to promote progression through S phase and enhance the stress resilience of neuroblastoma cells.


Subject(s)
N-Myc Proto-Oncogene Protein , Nuclear Proteins , Oncogene Proteins , RNA-Binding Proteins , N-Myc Proto-Oncogene Protein/metabolism , N-Myc Proto-Oncogene Protein/genetics , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Oncogene Proteins/metabolism , Oncogene Proteins/genetics , Promoter Regions, Genetic , Cell Line, Tumor , Neuroblastoma/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Exosomes/metabolism , Exosomes/genetics , Introns , Protein Binding , Cell Nucleus/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Gene Expression Regulation, Neoplastic , RNA/metabolism , RNA/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Cell Proliferation
10.
Cell ; 167(6): 1525-1539.e17, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912060

ABSTRACT

Poorly immunogenic tumor cells evade host immunity and grow even in the presence of an intact immune system, but the complex mechanisms regulating tumor immunogenicity have not been elucidated. Here, we discovered an unexpected role of the Hippo pathway in suppressing anti-tumor immunity. We demonstrate that, in three different murine syngeneic tumor models (B16, SCC7, and 4T1), loss of the Hippo pathway kinases LATS1/2 (large tumor suppressor 1 and 2) in tumor cells inhibits tumor growth. Tumor regression by LATS1/2 deletion requires adaptive immune responses, and LATS1/2 deficiency enhances tumor vaccine efficacy. Mechanistically, LATS1/2-null tumor cells secrete nucleic-acid-rich extracellular vesicles, which induce a type I interferon response via the Toll-like receptors-MYD88/TRIF pathway. LATS1/2 deletion in tumors thus improves tumor immunogenicity, leading to tumor destruction by enhancing anti-tumor immune responses. Our observations uncover a key role of the Hippo pathway in modulating tumor immunogenicity and demonstrate a proof of concept for targeting LATS1/2 in cancer immunotherapy.


Subject(s)
Immune Tolerance , Neoplasms/immunology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cancer Vaccines/immunology , Gene Deletion , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Toll-Like Receptors/metabolism , Tumor Suppressor Proteins/genetics
11.
Mol Cell ; 83(22): 4093-4105.e7, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37879335

ABSTRACT

The Ski2-Ski3-Ski8 (Ski238) helicase complex directs cytoplasmic mRNAs toward the nucleolytic exosome complex for degradation. In yeast, the interaction between Ski238 and exosome requires the adaptor protein Ski7. We determined different cryo-EM structures of the Ski238 complex depicting the transition from a rigid autoinhibited closed conformation to a flexible active open conformation in which the Ski2 helicase module has detached from the rest of Ski238. The open conformation favors the interaction of the Ski3 subunit with exosome-bound Ski7, leading to the recruitment of the exosome. In the Ski238-Ski7-exosome holocomplex, the Ski2 helicase module binds the exosome cap, enabling the RNA to traverse from the helicase through the internal exosome channel to the Rrp44 exoribonuclease. Our study pinpoints how conformational changes within the Ski238 complex regulate exosome recruitment for RNA degradation. We also reveal the remarkable conservation of helicase-exosome RNA channeling mechanisms throughout eukaryotic nuclear and cytoplasmic exosome complexes.


Subject(s)
Exosomes , Saccharomyces cerevisiae Proteins , Exosomes/metabolism , RNA/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , RNA Stability
12.
Mol Cell ; 83(13): 2222-2239.e5, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37329883

ABSTRACT

The transcriptional termination of unstable non-coding RNAs (ncRNAs) is poorly understood compared to coding transcripts. We recently identified ZC3H4-WDR82 ("restrictor") as restricting human ncRNA transcription, but how it does this is unknown. Here, we show that ZC3H4 additionally associates with ARS2 and the nuclear exosome targeting complex. The domains of ZC3H4 that contact ARS2 and WDR82 are required for ncRNA restriction, suggesting their presence in a functional complex. Consistently, ZC3H4, WDR82, and ARS2 co-transcriptionally control an overlapping population of ncRNAs. ZC3H4 is proximal to the negative elongation factor, PNUTS, which we show enables restrictor function and is required to terminate the transcription of all major RNA polymerase II transcript classes. In contrast to short ncRNAs, longer protein-coding transcription is supported by U1 snRNA, which shields transcripts from restrictor and PNUTS at hundreds of genes. These data provide important insights into the mechanism and control of transcription by restrictor and PNUTS.


Subject(s)
RNA Polymerase II , Transcription, Genetic , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Cell Nucleus/metabolism , RNA, Untranslated/genetics , Chromosomal Proteins, Non-Histone/genetics
13.
Mol Cell ; 82(4): 756-769.e8, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35120588

ABSTRACT

The superkiller (SKI) complex is the cytoplasmic co-factor and regulator of the RNA-degrading exosome. In human cells, the SKI complex functions mainly in co-translational surveillance-decay pathways, and its malfunction is linked to a severe congenital disorder, the trichohepatoenteric syndrome. To obtain insights into the molecular mechanisms regulating the human SKI (hSKI) complex, we structurally characterized several of its functional states in the context of 80S ribosomes and substrate RNA. In a prehydrolytic ATP form, the hSKI complex exhibits a closed conformation with an inherent gating system that effectively traps the 80S-bound RNA into the hSKI2 helicase subunit. When active, hSKI switches to an open conformation in which the gating is released and the RNA 3' end exits the helicase. The emerging picture is that the gatekeeping mechanism and architectural remodeling of hSKI underpin a regulated RNA channeling system that is mechanistically conserved among the cytoplasmic and nuclear helicase-exosome complexes.


Subject(s)
Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , RNA Helicases/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA/metabolism , Ribosome Subunits/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Exoribonucleases/genetics , Exoribonucleases/ultrastructure , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/ultrastructure , HEK293 Cells , Humans , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , RNA/genetics , RNA/ultrastructure , RNA Helicases/genetics , RNA Helicases/ultrastructure , Ribosome Subunits/genetics , Ribosome Subunits/ultrastructure , Structure-Activity Relationship
14.
Mol Cell ; 82(9): 1691-1707.e8, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35349793

ABSTRACT

Transposable elements (TEs) are widespread genetic parasites known to be kept under tight transcriptional control. Here, we describe a functional connection between the mouse-orthologous "nuclear exosome targeting" (NEXT) and "human silencing hub" (HUSH) complexes, involved in nuclear RNA decay and the epigenetic silencing of TEs, respectively. Knocking out the NEXT component ZCCHC8 in embryonic stem cells results in elevated TE RNA levels. We identify a physical interaction between ZCCHC8 and the MPP8 protein of HUSH and establish that HUSH recruits NEXT to chromatin at MPP8-bound TE loci. However, while NEXT and HUSH both dampen TE RNA expression, their activities predominantly affect shorter non-polyadenylated and full-length polyadenylated transcripts, respectively. Indeed, our data suggest that the repressive action of HUSH promotes a condition favoring NEXT RNA decay activity. In this way, transcriptional and post-transcriptional machineries synergize to suppress the genotoxic potential of TE RNAs.


Subject(s)
Exosome Multienzyme Ribonuclease Complex , Exosomes , Animals , Chromatin/genetics , Chromatin/metabolism , DNA Transposable Elements/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Exosomes/metabolism , Humans , Mice , Nuclear Proteins/metabolism , RNA/metabolism , RNA Stability
15.
Mol Cell ; 82(1): 159-176.e12, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34847357

ABSTRACT

The MYCN oncoprotein drives the development of numerous neuroendocrine and pediatric tumors. Here we show that MYCN interacts with the nuclear RNA exosome, a 3'-5' exoribonuclease complex, and recruits the exosome to its target genes. In the absence of the exosome, MYCN-directed elongation by RNA polymerase II (RNAPII) is slow and non-productive on a large group of cell-cycle-regulated genes. During the S phase of MYCN-driven tumor cells, the exosome is required to prevent the accumulation of stalled replication forks and of double-strand breaks close to the transcription start sites. Upon depletion of the exosome, activation of ATM causes recruitment of BRCA1, which stabilizes nuclear mRNA decapping complexes, leading to MYCN-dependent transcription termination. Disruption of mRNA decapping in turn activates ATR, indicating transcription-replication conflicts. We propose that exosome recruitment by MYCN maintains productive transcription elongation during S phase and prevents transcription-replication conflicts to maintain the rapid proliferation of neuroendocrine tumor cells.


Subject(s)
Cell Nucleus/enzymology , Cell Proliferation , DNA Replication , Exosomes/enzymology , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/enzymology , RNA Polymerase II/metabolism , Transcription, Genetic , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , DNA Breaks, Double-Stranded , Exoribonucleases/genetics , Exoribonucleases/metabolism , Exosomes/genetics , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice , N-Myc Proto-Oncogene Protein/genetics , NIH 3T3 Cells , Neuroblastoma/genetics , Neuroblastoma/pathology , Promoter Regions, Genetic , RNA Caps/genetics , RNA Caps/metabolism , RNA Polymerase II/genetics , Transcription Termination, Genetic
16.
Mol Cell ; 82(13): 2505-2518.e7, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35688157

ABSTRACT

In mammalian cells, spurious transcription results in a vast repertoire of unproductive non-coding RNAs, whose deleterious accumulation is prevented by rapid decay. The nuclear exosome targeting (NEXT) complex plays a central role in directing non-functional transcripts to exosome-mediated degradation, but the structural and molecular mechanisms remain enigmatic. Here, we elucidated the architecture of the human NEXT complex, showing that it exists as a dimer of MTR4-ZCCHC8-RBM7 heterotrimers. Dimerization preconfigures the major MTR4-binding region of ZCCHC8 and arranges the two MTR4 helicases opposite to each other, with each protomer able to function on many types of RNAs. In the inactive state of the complex, the 3' end of an RNA substrate is enclosed in the MTR4 helicase channel by a ZCCHC8 C-terminal gatekeeping domain. The architecture of a NEXT-exosome assembly points to the molecular and regulatory mechanisms with which the NEXT complex guides RNA substrates to the exosome.


Subject(s)
Exosomes , RNA , Cell Nucleus/genetics , Cell Nucleus/metabolism , DEAD-box RNA Helicases/metabolism , DNA Helicases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosomes/genetics , Exosomes/metabolism , Humans , Protein Binding , RNA/genetics , RNA/metabolism , RNA Helicases/metabolism , RNA Stability/genetics
17.
Mol Cell ; 81(2): 293-303.e4, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33326748

ABSTRACT

Ribosome assembly is catalyzed by numerous trans-acting factors and coupled with irreversible pre-rRNA processing, driving the pathway toward mature ribosomal subunits. One decisive step early in this progression is removal of the 5' external transcribed spacer (5'-ETS), an RNA extension at the 18S rRNA that is integrated into the huge 90S pre-ribosome structure. Upon endo-nucleolytic cleavage at an internal site, A1, the 5'-ETS is separated from the 18S rRNA and degraded. Here we present biochemical and cryo-electron microscopy analyses that depict the RNA exosome, a major 3'-5' exoribonuclease complex, in a super-complex with the 90S pre-ribosome. The exosome is docked to the 90S through its co-factor Mtr4 helicase, a processive RNA duplex-dismantling helicase, which strategically positions the exosome at the base of 5'-ETS helices H9-H9', which are dislodged in our 90S-exosome structures. These findings suggest a direct role of the exosome in structural remodeling of the 90S pre-ribosome to drive eukaryotic ribosome synthesis.


Subject(s)
DEAD-box RNA Helicases/chemistry , Endoribonucleases/chemistry , Exonucleases/chemistry , Exosome Multienzyme Ribonuclease Complex/ultrastructure , RNA, Ribosomal, 18S/chemistry , Ribosomes/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/genetics , Binding Sites , Cryoelectron Microscopy , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Exonucleases/genetics , Exonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Models, Molecular , Protein Binding , Protein Biosynthesis , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA Stability , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 18S/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
18.
Mol Cell ; 81(19): 3949-3964.e7, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34450044

ABSTRACT

Immunoglobulin heavy chain (IgH) locus-associated G-rich long noncoding RNA (SµGLT) is important for physiological and pathological B cell DNA recombination. We demonstrate that the METTL3 enzyme-catalyzed N6-methyladenosine (m6A) RNA modification drives recognition and 3' end processing of SµGLT by the RNA exosome, promoting class switch recombination (CSR) and suppressing chromosomal translocations. The recognition is driven by interaction of the MPP6 adaptor protein with nuclear m6A reader YTHDC1. MPP6 and YTHDC1 promote CSR by recruiting AID and the RNA exosome to actively transcribe SµGLT. Direct suppression of m6A modification of SµGLT or of m6A reader YTHDC1 reduces CSR. Moreover, METTL3, an essential gene for B cell development in the bone marrow and germinal center, suppresses IgH-associated aberrant DNA breaks and prevents genomic instability. Taken together, we propose coordinated and central roles for MPP6, m6A modification, and m6A reader proteins in controlling long noncoding RNA processing, DNA recombination, and development in B cells.


Subject(s)
Adenosine/analogs & derivatives , B-Lymphocytes/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Immunoglobulin Heavy Chains/metabolism , RNA 3' End Processing , RNA, Long Noncoding/metabolism , Recombination, Genetic , Adenosine/metabolism , Animals , B-Lymphocytes/immunology , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Female , Genomic Instability , HEK293 Cells , Humans , Immunoglobulin Class Switching , Immunoglobulin Heavy Chains/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mice, Knockout , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
19.
Mol Cell ; 77(6): 1340-1349.e6, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32006463

ABSTRACT

The evolutionarily conserved Ski2-Ski3-Ski8 (Ski) complex containing the 3'→5' RNA helicase Ski2 binds to 80S ribosomes near the mRNA entrance and facilitates 3'→5' exosomal degradation of mRNA during ribosome-associated mRNA surveillance pathways. Here, we assayed Ski's activity using an in vitro reconstituted translation system and report that this complex efficiently extracts mRNA from 80S ribosomes in the 3'→5' direction in a nucleotide-by-nucleotide manner. The process is ATP dependent and can occur on pre- and post-translocation ribosomal complexes. The Ski complex can engage productively with mRNA and extract it from 80S complexes containing as few as 19 (but not 13) 3'-terminal mRNA nucleotides starting from the P site. The mRNA-extracting activity of the Ski complex suggests that its role in mRNA quality control pathways is not limited to acceleration of exosomal degradation and could include clearance of stalled ribosomes from mRNA, poising mRNA for degradation and rendering stalled ribosomes recyclable by Pelota/Hbs1/ABCE1.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Exosomes/metabolism , GTP-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/isolation & purification , Ribosomes/metabolism , ATP-Binding Cassette Transporters/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Exosomes/genetics , GTP-Binding Proteins/genetics , Humans , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/genetics
20.
Immunol Rev ; 321(1): 169-180, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37950410

ABSTRACT

Cancer arises from the growth and division of uncontrolled erroneous cells. Programmed cell death (PCD), or regulated cell death (RCD), includes natural processes that eliminate damaged or abnormal cells. Dysregulation of PCD is a hallmark of cancer, as cancer cells often evade cell death and continue to proliferate. Exosomes nanoscale extracellular vesicles secreted by different types of cells carrying a variety of molecules, including nucleic acids, proteins, and lipids, to have indispensable role in the communication between cells, and can influence various cellular processes, including PCD. Exosomes have been shown to modulate PCD in cancer cells by transferring pro- or antideath molecules to neighboring cells. Additionally, exosomes can facilitate the spread of PCD to surrounding cancer cells, making them promising in the treatment of various cancers. The exosomes' diagnostic potential in cancer is also an active area of research. Exosomes can be isolated from a wide range of bodily fluids and tissues, such as blood and urine, and can provide a noninvasive way to monitor cancer progression and treatment response. Furthermore, exosomes have also been employed as a delivery system for therapeutic agents. By engineering exosomes to carry drugs or other therapeutic molecules, they can be targeted specifically to cancer cells, reducing toxicity to healthy tissues. Here, we discussed exosomes in the diagnosis and prevention of cancers, tumor immunotherapy, and drug delivery, as well as in different types of PCD.


Subject(s)
Exosomes , Extracellular Vesicles , Neoplasms , Humans , Exosomes/metabolism , Exosomes/pathology , Neoplasms/drug therapy , Extracellular Vesicles/pathology , Apoptosis
SELECTION OF CITATIONS
SEARCH DETAIL