Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 642
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 180(2): 278-295.e23, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31978345

RESUMEN

Mutations in FAMIN cause arthritis and inflammatory bowel disease in early childhood, and a common genetic variant increases the risk for Crohn's disease and leprosy. We developed an unbiased liquid chromatography-mass spectrometry screen for enzymatic activity of this orphan protein. We report that FAMIN phosphorolytically cleaves adenosine into adenine and ribose-1-phosphate. Such activity was considered absent from eukaryotic metabolism. FAMIN and its prokaryotic orthologs additionally have adenosine deaminase, purine nucleoside phosphorylase, and S-methyl-5'-thioadenosine phosphorylase activity, hence, combine activities of the namesake enzymes of central purine metabolism. FAMIN enables in macrophages a purine nucleotide cycle (PNC) between adenosine and inosine monophosphate and adenylosuccinate, which consumes aspartate and releases fumarate in a manner involving fatty acid oxidation and ATP-citrate lyase activity. This macrophage PNC synchronizes mitochondrial activity with glycolysis by balancing electron transfer to mitochondria, thereby supporting glycolytic activity and promoting oxidative phosphorylation and mitochondrial H+ and phosphate recycling.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Adenina/metabolismo , Adenosina/metabolismo , Adenosina Desaminasa/metabolismo , Cromatografía Liquida/métodos , Células HEK293 , Células Hep G2 , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Espectrometría de Masas/métodos , Enzimas Multifuncionales/genética , Fosforilación , Proteínas/genética , Nucleótidos de Purina/metabolismo , Purinas/metabolismo
2.
Cell ; 177(2): 384-398.e11, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30853218

RESUMEN

The signaling organelles of the innate immune system consist of oligomeric protein complexes known as supramolecular organizing centers (SMOCs). Examples of SMOCs include myddosomes and inflammasomes, which respectively induce transcription-dependent and -independent inflammatory responses. The common use of oligomeric structures as signaling platforms suggests multifunctionality, but each SMOC has a singular biochemically defined function. Here, we report that the myddosome is a multifunctional organizing center. In addition to promoting inflammatory transcription factor activation, the myddosome drives the rapid induction of glycolysis. We identify the kinase TBK1 as a myddosome component that promotes glycolysis, but not nuclear factor κB (NF-κB) activation. Synthetic immunology approaches further diversified SMOC activities, as we created interferon- or necroptosis-inducing myddosomes, inflammasomes that induce interferon responses instead of pyroptosis, and a SMOC-like nanomachine that induces interferon expression in response to a chemical ligand. These discoveries demonstrate the flexibility of immune signaling organelles, which permits the design of user-defined innate immune responses.


Asunto(s)
Inmunidad Innata/inmunología , Inmunidad Innata/fisiología , Transducción de Señal/inmunología , Animales , Glucólisis/inmunología , Inflamasomas , Ratones , Ratones Endogámicos C57BL , Enzimas Multifuncionales/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Orgánulos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Toll-Like
3.
Cell ; 178(2): 473-490.e26, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31230715

RESUMEN

We introduce APEX-seq, a method for RNA sequencing based on direct proximity labeling of RNA using the peroxidase enzyme APEX2. APEX-seq in nine distinct subcellular locales produced a nanometer-resolution spatial map of the human transcriptome as a resource, revealing extensive patterns of localization for diverse RNA classes and transcript isoforms. We uncover a radial organization of the nuclear transcriptome, which is gated at the inner surface of the nuclear pore for cytoplasmic export of processed transcripts. We identify two distinct pathways of messenger RNA localization to mitochondria, each associated with specific sets of transcripts for building complementary macromolecular machines within the organelle. APEX-seq should be widely applicable to many systems, enabling comprehensive investigations of the spatial transcriptome.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endonucleasas/metabolismo , Enzimas Multifuncionales/metabolismo , ARN/metabolismo , Análisis de Secuencia de ARN/métodos , Colorantes Fluorescentes/química , Células HEK293 , Humanos , Microscopía Fluorescente , Mitocondrias/genética , ARN/química , ARN Mensajero/química , ARN Mensajero/metabolismo , Transcriptoma
4.
Cell ; 169(3): 523-537.e15, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431250

RESUMEN

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.


Asunto(s)
Linfocitos B/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Núcleo Celular/metabolismo , ADN Helicasas/metabolismo , Exorribonucleasas/genética , Inestabilidad Genómica , Cadenas Pesadas de Inmunoglobulina/genética , Ratones , Enzimas Multifuncionales , Proteínas Nucleares/genética , ARN Helicasas , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/genética
5.
Mol Cell ; 84(16): 3044-3060.e11, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39142279

RESUMEN

G-quadruplexes (G4s) form throughout the genome and influence important cellular processes. Their deregulation can challenge DNA replication fork progression and threaten genome stability. Here, we demonstrate an unexpected role for the double-stranded DNA (dsDNA) translocase helicase-like transcription factor (HLTF) in responding to G4s. We show that HLTF, which is enriched at G4s in the human genome, can directly unfold G4s in vitro and uses this ATP-dependent translocase function to suppress G4 accumulation throughout the cell cycle. Additionally, MSH2 (a component of MutS heterodimers that bind G4s) and HLTF act synergistically to suppress G4 accumulation, restrict alternative lengthening of telomeres, and promote resistance to G4-stabilizing drugs. In a discrete but complementary role, HLTF restrains DNA synthesis when G4s are stabilized by suppressing primase-polymerase (PrimPol)-dependent repriming. Together, the distinct roles of HLTF in the G4 response prevent DNA damage and potentially mutagenic replication to safeguard genome stability.


Asunto(s)
ADN Primasa , Replicación del ADN , Proteínas de Unión al ADN , G-Cuádruplex , Inestabilidad Genómica , Proteína 2 Homóloga a MutS , Factores de Transcripción , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteína 2 Homóloga a MutS/genética , ADN Primasa/metabolismo , ADN Primasa/genética , Homeostasis del Telómero , Daño del ADN , Células HEK293 , Enzimas Multifuncionales/metabolismo , Enzimas Multifuncionales/genética , ADN Polimerasa Dirigida por ADN
6.
Mol Cell ; 83(20): 3692-3706.e5, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37832548

RESUMEN

The senataxin (SETX, Sen1 in yeasts) RNA-DNA hybrid resolving helicase regulates multiple nuclear transactions, including DNA replication, transcription, and DNA repair, but the molecular basis for Sen1 activities is ill defined. Here, Sen1 cryoelectron microscopy (cryo-EM) reconstructions reveal an elongated inchworm-like architecture. Sen1 is composed of an amino terminal helical repeat Sen1 N-terminal (Sen1N) regulatory domain that is flexibly linked to its C-terminal SF1B helicase motor core (Sen1Hel) via an intrinsically disordered tether. In an autoinhibited state, the Sen1Sen1N domain regulates substrate engagement by promoting occlusion of the RNA substrate-binding cleft. The X-ray structure of an activated Sen1Hel engaging single-stranded RNA and ADP-SO4 shows that the enzyme encircles RNA and implicates a single-nucleotide power stroke in the Sen1 RNA translocation mechanism. Together, our data unveil dynamic protein-protein and protein-RNA interfaces underpinning helicase regulation and inactivation of human SETX activity by RNA-binding-deficient mutants in ataxia with oculomotor apraxia 2 neurodegenerative disease.


Asunto(s)
Enfermedades Neurodegenerativas , ARN , Humanos , ARN/genética , Microscopía por Crioelectrón , ARN Helicasas/genética , ARN Helicasas/química , Enzimas Multifuncionales/genética , ADN/genética , Homeostasis , ADN Helicasas/genética
7.
Nature ; 613(7942): 187-194, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36544021

RESUMEN

R-loops are RNA-DNA-hybrid-containing nucleic acids with important cellular roles. Deregulation of R-loop dynamics can lead to DNA damage and genome instability1, which has been linked to the action of endonucleases such as XPG2-4. However, the mechanisms and cellular consequences of such processing have remained unclear. Here we identify a new population of RNA-DNA hybrids in the cytoplasm that are R-loop-processing products. When nuclear R-loops were perturbed by depleting the RNA-DNA helicase senataxin (SETX) or the breast cancer gene BRCA1 (refs. 5-7), we observed XPG- and XPF-dependent cytoplasmic hybrid formation. We identify their source as a subset of stable, overlapping nuclear hybrids with a specific nucleotide signature. Cytoplasmic hybrids bind to the pattern recognition receptors cGAS and TLR3 (ref. 8), activating IRF3 and inducing apoptosis. Excised hybrids and an R-loop-induced innate immune response were also observed in SETX-mutated cells from patients with ataxia oculomotor apraxia type 2 (ref. 9) and in BRCA1-mutated cancer cells10. These findings establish RNA-DNA hybrids as immunogenic species that aberrantly accumulate in the cytoplasm after R-loop processing, linking R-loop accumulation to cell death through the innate immune response. Aberrant R-loop processing and subsequent innate immune activation may contribute to many diseases, such as neurodegeneration and cancer.


Asunto(s)
Citoplasma , ADN , Reconocimiento de Inmunidad Innata , Ácidos Nucleicos Heterodúplex , Estructuras R-Loop , ARN , Humanos , Apoptosis , Citoplasma/inmunología , Citoplasma/metabolismo , ADN/química , ADN/inmunología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Genes BRCA1 , Enzimas Multifuncionales/genética , Enzimas Multifuncionales/metabolismo , Mutación , Neoplasias , Ácidos Nucleicos Heterodúplex/química , Ácidos Nucleicos Heterodúplex/inmunología , Estructuras R-Loop/inmunología , ARN/química , ARN/inmunología , ARN Helicasas/genética , ARN Helicasas/metabolismo , Ataxias Espinocerebelosas/genética
8.
Mol Cell ; 81(19): 4008-4025.e7, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34508659

RESUMEN

BRCA1/2 mutant tumor cells display an elevated mutation burden, the etiology of which remains unclear. Here, we report that these cells accumulate ssDNA gaps and spontaneous mutations during unperturbed DNA replication due to repriming by the DNA primase-polymerase PRIMPOL. Gap accumulation requires the DNA glycosylase SMUG1 and is exacerbated by depletion of the translesion synthesis (TLS) factor RAD18 or inhibition of the error-prone TLS polymerase complex REV1-Polζ by the small molecule JH-RE-06. JH-RE-06 treatment of BRCA1/2-deficient cells results in reduced mutation rates and PRIMPOL- and SMUG1-dependent loss of viability. Through cellular and animal studies, we demonstrate that JH-RE-06 is preferentially toxic toward HR-deficient cancer cells. Furthermore, JH-RE-06 remains effective toward PARP inhibitor (PARPi)-resistant BRCA1 mutant cells and displays additive toxicity with crosslinking agents or PARPi. Collectively, these studies identify a protective and mutagenic role for REV1-Polζ in BRCA1/2 mutant cells and provide the rationale for using REV1-Polζ inhibitors to treat BRCA1/2 mutant tumors.


Asunto(s)
Roturas del ADN de Cadena Simple , ADN Primasa/metabolismo , Replicación del ADN , ADN de Neoplasias/biosíntesis , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Enzimas Multifuncionales/metabolismo , Neoplasias/enzimología , Nucleotidiltransferasas/metabolismo , Reparación del ADN por Recombinación , Animales , Antineoplásicos/farmacología , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Línea Celular Tumoral , ADN Primasa/genética , ADN de Neoplasias/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Femenino , Células HEK293 , Humanos , Ratones Desnudos , Enzimas Multifuncionales/genética , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/genética , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Cell ; 81(19): 4026-4040.e8, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34624216

RESUMEN

PRIMPOL repriming allows DNA replication to skip DNA lesions, leading to ssDNA gaps. These gaps must be filled to preserve genome stability. Using a DNA fiber approach to directly monitor gap filling, we studied the post-replicative mechanisms that fill the ssDNA gaps generated in cisplatin-treated cells upon increased PRIMPOL expression or when replication fork reversal is defective because of SMARCAL1 inactivation or PARP inhibition. We found that a mechanism dependent on the E3 ubiquitin ligase RAD18, PCNA monoubiquitination, and the REV1 and POLζ translesion synthesis polymerases promotes gap filling in G2. The E2-conjugating enzyme UBC13, the RAD51 recombinase, and REV1-POLζ are instead responsible for gap filling in S, suggesting that temporally distinct pathways of gap filling operate throughout the cell cycle. Furthermore, we found that BRCA1 and BRCA2 promote gap filling by limiting MRE11 activity and that simultaneously targeting fork reversal and gap filling enhances chemosensitivity in BRCA-deficient cells.


Asunto(s)
Roturas del ADN de Cadena Simple , ADN Primasa/metabolismo , Reparación del ADN , Replicación del ADN , ADN de Neoplasias/biosíntesis , ADN Polimerasa Dirigida por ADN/metabolismo , Fase G2 , Enzimas Multifuncionales/metabolismo , Neoplasias/metabolismo , Fase S , Antineoplásicos/farmacología , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Línea Celular Tumoral , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Primasa/genética , ADN de Neoplasias/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Inestabilidad Genómica , Células HEK293 , Humanos , Proteína Homóloga de MRE11/genética , Proteína Homóloga de MRE11/metabolismo , Enzimas Multifuncionales/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Tiempo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
10.
Genes Dev ; 35(23-24): 1579-1594, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34819354

RESUMEN

The nucleolus is an important cellular compartment in which ribosomal RNAs (rRNAs) are transcribed and where certain stress pathways that are crucial for cell growth are coordinated. Here we report novel functions of the DNA replication and repair factor replication protein A (RPA) in control of nucleolar homeostasis. We show that loss of the DNA:RNA helicase senataxin (SETX) promotes RPA nucleolar localization, and that this relocalization is dependent on the presence of R loops. Notably, this nucleolar RPA phenotype was also observed in the presence of camptothecin (CPT)-induced genotoxic stress, as well as in SETX-deficient AOA2 patient fibroblasts. Extending these results, we found that RPA is recruited to rDNA following CPT treatment, where RPA prevents R-loop-induced DNA double-strand breaks. Furthermore, we show that loss of RPA significantly decreased 47S pre-rRNA levels, which was accompanied by increased expression of both RNAP II-mediated "promoter and pre-rRNA antisense" RNA as well as RNAP I-transcribed intragenic spacer RNAs. Finally, and likely reflecting the above, we found that loss of RPA promoted nucleolar structural disorganization, characterized by the appearance of reduced size nucleoli. Our findings both indicate new roles for RPA in nucleoli through pre-rRNA transcriptional control and also emphasize that RPA function in nucleolar homeostasis is linked to R-loop resolution under both physiological and pathological conditions.


Asunto(s)
Estructuras R-Loop , Proteína de Replicación A , Nucléolo Celular/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Humanos , Enzimas Multifuncionales , ARN Helicasas/metabolismo , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , Transcripción Genética
11.
Nature ; 606(7916): 945-952, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35732742

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a heterogenous neurodegenerative disorder that affects motor neurons and voluntary muscle control1. ALS heterogeneity includes the age of manifestation, the rate of progression and the anatomical sites of symptom onset. Disease-causing mutations in specific genes have been identified and define different subtypes of ALS1. Although several ALS-associated genes have been shown to affect immune functions2, whether specific immune features account for ALS heterogeneity is poorly understood. Amyotrophic lateral sclerosis-4 (ALS4) is characterized by juvenile onset and slow progression3. Patients with ALS4 show motor difficulties by the time that they are in their thirties, and most of them require devices to assist with walking by their fifties. ALS4 is caused by mutations in the senataxin gene (SETX). Here, using Setx knock-in mice that carry the ALS4-causative L389S mutation, we describe an immunological signature that consists of clonally expanded, terminally differentiated effector memory (TEMRA) CD8 T cells in the central nervous system and the blood of knock-in mice. Increased frequencies of antigen-specific CD8 T cells in knock-in mice mirror the progression of motor neuron disease and correlate with anti-glioma immunity. Furthermore, bone marrow transplantation experiments indicate that the immune system has a key role in ALS4 neurodegeneration. In patients with ALS4, clonally expanded TEMRA CD8 T cells circulate in the peripheral blood. Our results provide evidence of an antigen-specific CD8 T cell response in ALS4, which could be used to unravel disease mechanisms and as a potential biomarker of disease state.


Asunto(s)
Esclerosis Amiotrófica Lateral , Linfocitos T CD8-positivos , Células Clonales , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/patología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Células Clonales/patología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Técnicas de Sustitución del Gen , Ratones , Neuronas Motoras/patología , Enzimas Multifuncionales/genética , Enzimas Multifuncionales/metabolismo , Mutación , ARN Helicasas/genética , ARN Helicasas/metabolismo
12.
Mol Cell ; 78(6): 1152-1165.e8, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32516598

RESUMEN

The APEX2 gene encodes APE2, a nuclease related to APE1, the apurinic/apyrimidinic endonuclease acting in base excision repair. Loss of APE2 is lethal in cells with mutated BRCA1 or BRCA2, making APE2 a prime target for homologous recombination-defective cancers. However, because the function of APE2 in DNA repair is poorly understood, it is unclear why BRCA-deficient cells require APE2 for viability. Here we present the genetic interaction profiles of APE2, APE1, and TDP1 deficiency coupled to biochemical and structural dissection of APE2. We conclude that the main role of APE2 is to reverse blocked 3' DNA ends, problematic lesions that preclude DNA synthesis. Our work also suggests that TOP1 processing of genomic ribonucleotides is the main source of 3'-blocking lesions relevant to APEX2-BRCA1/2 synthetic lethality. The exquisite sensitivity of BRCA-deficient cells to 3' blocks indicates that they represent a tractable vulnerability in homologous recombination-deficient tumor cells.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endonucleasas/metabolismo , Enzimas Multifuncionales/metabolismo , Proteína BRCA1/genética , Proteína BRCA2/genética , Línea Celular , ADN/metabolismo , Daño del ADN , Reparación del ADN/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Endonucleasas/genética , Genes BRCA1/fisiología , Humanos , Enzimas Multifuncionales/genética , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo
13.
Mol Cell ; 78(6): 1237-1251.e7, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32442397

RESUMEN

DNA replication stress can stall replication forks, leading to genome instability. DNA damage tolerance pathways assist fork progression, promoting replication fork reversal, translesion DNA synthesis (TLS), and repriming. In the absence of the fork remodeler HLTF, forks fail to slow following replication stress, but underlying mechanisms and cellular consequences remain elusive. Here, we demonstrate that HLTF-deficient cells fail to undergo fork reversal in vivo and rely on the primase-polymerase PRIMPOL for repriming, unrestrained replication, and S phase progression upon limiting nucleotide levels. By contrast, in an HLTF-HIRAN mutant, unrestrained replication relies on the TLS protein REV1. Importantly, HLTF-deficient cells also exhibit reduced double-strand break (DSB) formation and increased survival upon replication stress. Our findings suggest that HLTF promotes fork remodeling, preventing other mechanisms of replication stress tolerance in cancer cells. This remarkable plasticity of the replication fork may determine the outcome of replication stress in terms of genome integrity, tumorigenesis, and response to chemotherapy.


Asunto(s)
Replicación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , ADN/biosíntesis , Factores de Transcripción/metabolismo , Línea Celular Tumoral , ADN/genética , Daño del ADN/genética , ADN Primasa/metabolismo , ADN Primasa/fisiología , Reparación del ADN/genética , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , ADN Polimerasa Dirigida por ADN/fisiología , Células HEK293 , Humanos , Células K562 , Enzimas Multifuncionales/metabolismo , Enzimas Multifuncionales/fisiología , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/fisiología , Factores de Transcripción/genética
14.
Mol Cell ; 77(3): 461-474.e9, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31676232

RESUMEN

Acute treatment with replication-stalling chemotherapeutics causes reversal of replication forks. BRCA proteins protect reversed forks from nucleolytic degradation, and their loss leads to chemosensitivity. Here, we show that fork degradation is no longer detectable in BRCA1-deficient cancer cells exposed to multiple cisplatin doses, mimicking a clinical treatment regimen. This effect depends on increased expression and chromatin loading of PRIMPOL and is regulated by ATR activity. Electron microscopy and single-molecule DNA fiber analyses reveal that PRIMPOL rescues fork degradation by reinitiating DNA synthesis past DNA lesions. PRIMPOL repriming leads to accumulation of ssDNA gaps while suppressing fork reversal. We propose that cells adapt to repeated cisplatin doses by activating PRIMPOL repriming under conditions that would otherwise promote pathological reversed fork degradation. This effect is generalizable to other conditions of impaired fork reversal (e.g., SMARCAL1 loss or PARP inhibition) and suggests a new strategy to modulate cisplatin chemosensitivity by targeting the PRIMPOL pathway.


Asunto(s)
ADN Primasa/metabolismo , Replicación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/metabolismo , Enzimas Multifuncionales/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , ADN/genética , Daño del ADN/genética , Daño del ADN/fisiología , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Primasa/fisiología , Replicación del ADN/genética , Replicación del ADN/fisiología , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/fisiología , Células HEK293 , Humanos , Enzimas Multifuncionales/fisiología , Ubiquitina-Proteína Ligasas/genética
15.
Mol Cell ; 78(3): 493-505.e8, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32353257

RESUMEN

The promyelocytic leukemia (PML) body is a phase-separated nuclear structure physically associated with chromatin, implying its crucial roles in genome functions. However, its role in transcriptional regulation is largely unknown. We developed APEX-mediated chromatin labeling and purification (ALaP) to identify the genomic regions proximal to PML bodies. We found that PML bodies associate with active regulatory regions across the genome and with ∼300 kb of the short arm of the Y chromosome (YS300) in mouse embryonic stem cells. The PML body association with YS300 is essential for the transcriptional activity of the neighboring Y-linked clustered genes. Mechanistically, PML bodies provide specific nuclear spaces that the de novo DNA methyltransferase DNMT3A cannot access, resulting in the steady maintenance of a hypo-methylated state at Y-linked gene promoters. Our study underscores a new mechanism for gene regulation in the 3D nuclear space and provides insights into the functional properties of nuclear structures for genome function.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Regulación de la Expresión Génica , Cuerpos de Inclusión Intranucleares/genética , Cromosoma Y/genética , Animales , Línea Celular , Cromatina/genética , Cromatina/metabolismo , ARN Helicasas DEAD-box/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Células Madre Embrionarias/fisiología , Endonucleasas/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Cuerpos de Inclusión Intranucleares/metabolismo , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/genética , Enzimas Multifuncionales/genética , Familia de Multigenes , Estrés Oxidativo , Proteína de la Leucemia Promielocítica/genética , Proteína de la Leucemia Promielocítica/metabolismo , Proteínas/genética , Factores de Transcripción/genética , Cromosoma Y/metabolismo
16.
Nat Immunol ; 16(5): 485-94, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25822250

RESUMEN

The human helicase senataxin (SETX) has been linked to the neurodegenerative diseases amyotrophic lateral sclerosis (ALS4) and ataxia with oculomotor apraxia (AOA2). Here we identified a role for SETX in controlling the antiviral response. Cells that had undergone depletion of SETX and SETX-deficient cells derived from patients with AOA2 had higher expression of antiviral mediators in response to infection than did wild-type cells. Mechanistically, we propose a model whereby SETX attenuates the activity of RNA polymerase II (RNAPII) at genes stimulated after a virus is sensed and thus controls the magnitude of the host response to pathogens and the biogenesis of various RNA viruses (e.g., influenza A virus and West Nile virus). Our data indicate a potentially causal link among inborn errors in SETX, susceptibility to infection and the development of neurologic disorders.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Gripe Humana/inmunología , Orthomyxoviridae/fisiología , ARN Helicasas/metabolismo , ARN Polimerasa II/metabolismo , Degeneraciones Espinocerebelosas/genética , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Citocinas/metabolismo , ADN Helicasas , Perros , Regulación hacia Abajo , Humanos , Inmunidad Innata/genética , Factor 3 Regulador del Interferón/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Análisis por Micromatrices , Enzimas Multifuncionales , ARN Helicasas/genética , ARN Polimerasa II/genética , ARN Interferente Pequeño/genética , Ataxias Espinocerebelosas/congénito , Células Vero , Replicación Viral/genética
17.
Cell ; 150(6): 1147-57, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22980978

RESUMEN

Transcription elongation is increasingly recognized as an important mechanism of gene regulation. Here, we show that microprocessor controls gene expression in an RNAi-independent manner. Microprocessor orchestrates the recruitment of termination factors Setx and Xrn2, and the 3'-5' exoribonuclease, Rrp6, to initiate RNAPII pausing and premature termination at the HIV-1 promoter through cleavage of the stem-loop RNA, TAR. Rrp6 further processes the cleavage product, which generates a small RNA that is required to mediate potent transcriptional repression and chromatin remodeling at the HIV-1 promoter. Using chromatin immunoprecipitation coupled to high-throughput sequencing (ChIP-seq), we identified cellular gene targets whose transcription is modulated by microprocessor. Our study reveals RNAPII pausing and premature termination mediated by the co-operative activity of ribonucleases, Drosha/Dgcr8, Xrn2, and Rrp6, as a regulatory mechanism of RNAPII-dependent transcription elongation.


Asunto(s)
Exorribonucleasas/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Regulación Viral de la Expresión Génica , VIH-1/genética , ARN Helicasas/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética , Secuencia de Bases , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , ADN Helicasas , Duplicado del Terminal Largo de VIH , Humanos , Datos de Secuencia Molecular , Enzimas Multifuncionales , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Viral/química , ARN Viral/genética , Factores de Transcripción/metabolismo
18.
Mol Cell ; 75(4): 875-887.e5, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442426

RESUMEN

Diverse ribonucleoprotein complexes control mRNA processing, translation, and decay. Transcripts in these complexes localize to specific regions of the cell and can condense into non-membrane-bound structures such as stress granules. It has proven challenging to map the RNA composition of these large and dynamic structures, however. We therefore developed an RNA proximity labeling technique, APEX-seq, which uses the ascorbate peroxidase APEX2 to probe the spatial organization of the transcriptome. We show that APEX-seq can resolve the localization of RNAs within the cell and determine their enrichment or depletion near key RNA-binding proteins. Matching the spatial transcriptome, as revealed by APEX-seq, with the spatial proteome determined by APEX-mass spectrometry (APEX-MS), obtained precisely in parallel, provides new insights into the organization of translation initiation complexes on active mRNAs and unanticipated complexity in stress granule composition. Our novel technique allows a powerful and general approach to explore the spatial environment of macromolecules.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endonucleasas/metabolismo , Enzimas Multifuncionales/metabolismo , Iniciación de la Cadena Peptídica Traduccional , ARN/metabolismo , Coloración y Etiquetado , Transcriptoma , Gránulos Citoplasmáticos/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Endonucleasas/genética , Células HEK293 , Humanos , Enzimas Multifuncionales/genética , ARN/genética
19.
Mol Cell ; 73(5): 885-899.e6, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30686591

RESUMEN

BRCA1 or BRCA2 inactivation drives breast and ovarian cancer but also creates vulnerability to poly(ADP-ribose) polymerase (PARP) inhibitors. To search for additional targets whose inhibition is synthetically lethal in BRCA2-deficient backgrounds, we screened two pairs of BRCA2 isogenic cell lines with DNA-repair-focused small hairpin RNA (shRNA) and CRISPR (clustered regularly interspaced short palindromic repeats)-based libraries. We found that BRCA2-deficient cells are selectively dependent on multiple pathways including base excision repair, ATR signaling, and splicing. We identified APEX2 and FEN1 as synthetic lethal genes with both BRCA1 and BRCA2 loss of function. BRCA2-deficient cells require the apurinic endonuclease activity and the PCNA-binding domain of Ape2 (APEX2), but not Ape1 (APEX1). Furthermore, BRCA2-deficient cells require the 5' flap endonuclease but not the 5'-3' exonuclease activity of Fen1, and chemically inhibiting Fen1 selectively targets BRCA-deficient cells. Finally, we developed a microhomology-mediated end-joining (MMEJ) reporter and showed that Fen1 participates in MMEJ, underscoring the importance of MMEJ as a collateral repair pathway in the context of homologous recombination (HR) deficiency.


Asunto(s)
Proteína BRCA2/genética , Sistemas CRISPR-Cas , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Endonucleasas de ADN Solapado/genética , Genes Letales , Neoplasias/genética , Interferencia de ARN , Mutaciones Letales Sintéticas , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Muerte Celular , Línea Celular Tumoral , Daño del ADN , Reparación del ADN por Unión de Extremidades , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endonucleasas , Endonucleasas de ADN Solapado/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Enzimas Multifuncionales , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Interferente Pequeño/genética
20.
Proc Natl Acad Sci U S A ; 121(16): e2322924121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38607933

RESUMEN

Many Mendelian disorders, such as Huntington's disease (HD) and spinocerebellar ataxias, arise from expansions of CAG trinucleotide repeats. Despite the clear genetic causes, additional genetic factors may influence the rate of those monogenic disorders. Notably, genome-wide association studies discovered somewhat expected modifiers, particularly mismatch repair genes involved in the CAG repeat instability, impacting age at onset of HD. Strikingly, FAN1, previously unrelated to repeat instability, produced the strongest HD modification signals. Diverse FAN1 haplotypes independently modify HD, with rare genetic variants diminishing DNA binding or nuclease activity of the FAN1 protein, hastening HD onset. However, the mechanism behind the frequent and the most significant onset-delaying FAN1 haplotype lacking missense variations has remained elusive. Here, we illustrated that a microRNA acting on 3'-UTR (untranslated region) SNP rs3512, rather than transcriptional regulation, is responsible for the significant FAN1 expression quantitative trait loci signal and allelic imbalance in FAN1 messenger ribonucleic acid (mRNA), accounting for the most significant and frequent onset-delaying modifier haplotype in HD. Specifically, miR-124-3p selectively targets the reference allele at rs3512, diminishing the stability of FAN1 mRNA harboring that allele and consequently reducing its levels. Subsequent validation analyses, including the use of antagomir and 3'-UTR reporter vectors with swapped alleles, confirmed the specificity of miR-124-3p at rs3512. Together, these findings indicate that the alternative allele at rs3512 renders the FAN1 mRNA less susceptible to miR-124-3p-mediated posttranscriptional regulation, resulting in increased FAN1 levels and a subsequent delay in HD onset by mitigating CAG repeat instability.


Asunto(s)
Enfermedad de Huntington , MicroARNs , Humanos , Regiones no Traducidas 3'/genética , Endodesoxirribonucleasas , Exodesoxirribonucleasas/genética , Estudio de Asociación del Genoma Completo , Enfermedad de Huntington/genética , MicroARNs/genética , Enzimas Multifuncionales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA