Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 81(4): 859-869.e8, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33352108

RESUMEN

Active DNA demethylation via ten-eleven translocation (TET) family enzymes is essential for epigenetic reprogramming in cell state transitions. TET enzymes catalyze up to three successive oxidations of 5-methylcytosine (5mC), generating 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), or 5-carboxycytosine (5caC). Although these bases are known to contribute to distinct demethylation pathways, the lack of tools to uncouple these sequential oxidative events has constrained our mechanistic understanding of the role of TETs in chromatin reprogramming. Here, we describe the first application of biochemically engineered TET mutants that unlink 5mC oxidation steps, examining their effects on somatic cell reprogramming. We show that only TET enzymes proficient for oxidation to 5fC/5caC can rescue the reprogramming potential of Tet2-deficient mouse embryonic fibroblasts. This effect correlated with rapid DNA demethylation at reprogramming enhancers and increased chromatin accessibility later in reprogramming. These experiments demonstrate that DNA demethylation through 5fC/5caC has roles distinct from 5hmC in somatic reprogramming to pluripotency.


Asunto(s)
5-Metilcitosina/metabolismo , Reprogramación Celular , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/metabolismo , Elementos de Facilitación Genéticos , Epigénesis Genética , Fibroblastos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas de Unión al ADN/genética , Dioxigenasas , Embrión de Mamíferos/citología , Fibroblastos/citología , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Mutación , Células 3T3 NIH , Proteínas Proto-Oncogénicas/genética
2.
Nucleic Acids Res ; 52(7): e38, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38407446

RESUMEN

The Infinium BeadChip is the most widely used DNA methylome assay technology for population-scale epigenome profiling. However, the standard workflow requires over 200 ng of input DNA, hindering its application to small cell-number samples, such as primordial germ cells. We developed experimental and analysis workflows to extend this technology to suboptimal input DNA conditions, including ultra-low input down to single cells. DNA preamplification significantly enhanced detection rates to over 50% in five-cell samples and ∼25% in single cells. Enzymatic conversion also substantially improved data quality. Computationally, we developed a method to model the background signal's influence on the DNA methylation level readings. The modified detection P-value calculation achieved higher sensitivities for low-input datasets and was validated in over 100 000 public diverse methylome profiles. We employed the optimized workflow to query the demethylation dynamics in mouse primordial germ cells available at low cell numbers. Our data revealed nuanced chromatin states, sex disparities, and the role of DNA methylation in transposable element regulation during germ cell development. Collectively, we present comprehensive experimental and computational solutions to extend this widely used methylation assay technology to applications with limited DNA.


Asunto(s)
Metilación de ADN , Análisis de la Célula Individual , Animales , Femenino , Humanos , Masculino , Ratones , Islas de CpG , ADN/genética , ADN/metabolismo , Epigenómica/métodos , Células Germinativas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de la Célula Individual/métodos
3.
Nucleic Acids Res ; 52(4): 2078-2090, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38261989

RESUMEN

The partnership of DNA deaminase enzymes with CRISPR-Cas nucleases is now a well-established method to enable targeted genomic base editing. However, an understanding of how Cas9 and DNA deaminases collaborate to shape base editor (BE) outcomes has been lacking. Here, we support a novel mechanistic model of base editing by deriving a range of hyperactive activation-induced deaminase (AID) base editors (hBEs) and exploiting their characteristic diversifying activity. Our model involves multiple layers of previously underappreciated cooperativity in BE steps including: (i) Cas9 binding can potentially expose both DNA strands for 'capture' by the deaminase, a feature that is enhanced by guide RNA mismatches; (ii) after strand capture, the intrinsic activity of the DNA deaminase can tune window size and base editing efficiency; (iii) Cas9 defines the boundaries of editing on each strand, with deamination blocked by Cas9 binding to either the PAM or the protospacer and (iv) non-canonical edits on the guide RNA bound strand can be further elicited by changing which strand is nicked by Cas9. Leveraging insights from our mechanistic model, we create novel hBEs that can remarkably generate simultaneous C > T and G > A transitions over >65 bp with significant potential for targeted gene diversification.


Asunto(s)
Proteína 9 Asociada a CRISPR , Citidina Desaminasa , Escherichia coli , Edición Génica , Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas , Citidina Desaminasa/metabolismo , ADN/genética , Escherichia coli/metabolismo , Mutación , ARN Guía de Sistemas CRISPR-Cas , Humanos , Animales
4.
Proc Natl Acad Sci U S A ; 120(36): e2303859120, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37639593

RESUMEN

Recurrent chromosomal rearrangements found in rhabdomyosarcoma (RMS) produce the PAX3-FOXO1 fusion protein, which is an oncogenic driver and a dependency in this disease. One important function of PAX3-FOXO1 is to arrest myogenic differentiation, which is linked to the ability of RMS cells to gain an unlimited proliferation potential. Here, we developed a phenotypic screening strategy for identifying factors that collaborate with PAX3-FOXO1 to block myo-differentiation in RMS. Unlike most genes evaluated in our screen, we found that loss of any of the three subunits of the Nuclear Factor Y (NF-Y) complex leads to a myo-differentiation phenotype that resembles the effect of inactivating PAX3-FOXO1. While the transcriptomes of NF-Y- and PAX3-FOXO1-deficient RMS cells bear remarkable similarity to one another, we found that these two transcription factors occupy nonoverlapping sites along the genome: NF-Y preferentially occupies promoters, whereas PAX3-FOXO1 primarily binds to distal enhancers. By integrating multiple functional approaches, we map the PAX3 promoter as the point of intersection between these two regulators. We show that NF-Y occupies CCAAT motifs present upstream of PAX3 to function as a transcriptional activator of PAX3-FOXO1 expression in RMS. These findings reveal a critical upstream role of NF-Y in the oncogenic PAX3-FOXO1 pathway, highlighting how a broadly essential transcription factor can perform tumor-specific roles in governing cellular state.


Asunto(s)
Rabdomiosarcoma , Factor de Unión a CCAAT/genética , Diferenciación Celular/genética , Aberraciones Cromosómicas , Rabdomiosarcoma/genética , Factores de Transcripción
5.
Nat Chem Biol ; 19(8): 1004-1012, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37322153

RESUMEN

5-methylcytosine (5mC) is the most important DNA modification in mammalian genomes. The ideal method for 5mC localization would be both nondestructive of DNA and direct, without requiring inference based on detection of unmodified cytosines. Here we present direct methylation sequencing (DM-Seq), a bisulfite-free method for profiling 5mC at single-base resolution using nanogram quantities of DNA. DM-Seq employs two key DNA-modifying enzymes: a neomorphic DNA methyltransferase and a DNA deaminase capable of precise discrimination between cytosine modification states. Coupling these activities with deaminase-resistant adapters enables accurate detection of only 5mC via a C-to-T transition in sequencing. By comparison, we uncover a PCR-related underdetection bias with the hybrid enzymatic-chemical TET-assisted pyridine borane sequencing approach. Importantly, we show that DM-Seq, unlike bisulfite sequencing, unmasks prognostically important CpGs in a clinical tumor sample by not confounding 5mC with 5-hydroxymethylcytosine. DM-Seq thus offers an all-enzymatic, nondestructive, faithful and direct method for the reading of 5mC alone.


Asunto(s)
5-Metilcitosina , Metilación de ADN , Animales , Citosina , ADN/genética , Análisis de Secuencia de ADN/métodos , Mamíferos/genética
6.
Nature ; 569(7757): 581-585, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31043749

RESUMEN

Methylation of cytosine to 5-methylcytosine (5mC) is a prevalent DNA modification found in many organisms. Sequential oxidation of 5mC by ten-eleven translocation (TET) dioxygenases results in a cascade of additional epigenetic marks and promotes demethylation of DNA in mammals1,2. However, the enzymatic activity and function of TET homologues in other eukaryotes remains largely unexplored. Here we show that the green alga Chlamydomonas reinhardtii contains a 5mC-modifying enzyme (CMD1) that is a TET homologue and catalyses the conjugation of a glyceryl moiety to the methyl group of 5mC through a carbon-carbon bond, resulting in two stereoisomeric nucleobase products. The catalytic activity of CMD1 requires Fe(II) and the integrity of its binding motif His-X-Asp, which is conserved in Fe-dependent dioxygenases3. However, unlike previously described TET enzymes, which use 2-oxoglutarate as a co-substrate4, CMD1 uses L-ascorbic acid (vitamin C) as an essential co-substrate. Vitamin C donates the glyceryl moiety to 5mC with concurrent formation of glyoxylic acid and CO2. The vitamin-C-derived DNA modification is present in the genome of wild-type C. reinhardtii but at a substantially lower level in a CMD1 mutant strain. The fitness of CMD1 mutant cells during exposure to high light levels is reduced. LHCSR3, a gene that is critical for the protection of C. reinhardtii from photo-oxidative damage under high light conditions, is hypermethylated and downregulated in CMD1 mutant cells compared to wild-type cells, causing a reduced capacity for photoprotective non-photochemical quenching. Our study thus identifies a eukaryotic DNA base modification that is catalysed by a divergent TET homologue and unexpectedly derived from vitamin C, and describes its role as a potential epigenetic mark that may counteract DNA methylation in the regulation of photosynthesis.


Asunto(s)
5-Metilcitosina/metabolismo , Proteínas Algáceas/metabolismo , Ácido Ascórbico/metabolismo , Biocatálisis , Chlamydomonas reinhardtii/enzimología , ADN/química , ADN/metabolismo , 5-Metilcitosina/química , Dióxido de Carbono/metabolismo , Metilación de ADN , Glioxilatos/metabolismo , Nucleósidos/química , Nucleósidos/metabolismo , Fotosíntesis
7.
Nature ; 558(7709): 307-312, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29849141

RESUMEN

Cancer immunotherapy based on genetically redirecting T cells has been used successfully to treat B cell malignancies1-3. In this strategy, the T cell genome is modified by integration of viral vectors or transposons encoding chimaeric antigen receptors (CARs) that direct tumour cell killing. However, this approach is often limited by the extent of expansion and persistence of CAR T cells4,5. Here we report mechanistic insights from studies of a patient with chronic lymphocytic leukaemia treated with CAR T cells targeting the CD19 protein. Following infusion of CAR T cells, anti-tumour activity was evident in the peripheral blood, lymph nodes and bone marrow; this activity was accompanied by complete remission. Unexpectedly, at the peak of the response, 94% of CAR T cells originated from a single clone in which lentiviral vector-mediated insertion of the CAR transgene disrupted the methylcytosine dioxygenase TET2 gene. Further analysis revealed a hypomorphic mutation in this patient's second TET2 allele. TET2-disrupted CAR T cells exhibited an epigenetic profile consistent with altered T cell differentiation and, at the peak of expansion, displayed a central memory phenotype. Experimental knockdown of TET2 recapitulated the potency-enhancing effect of TET2 dysfunction in this patient's CAR T cells. These findings suggest that the progeny of a single CAR T cell induced leukaemia remission and that TET2 modification may be useful for improving immunotherapies.


Asunto(s)
5-Metilcitosina/metabolismo , Antígenos CD19/inmunología , Dioxigenasas/genética , Inmunoterapia/métodos , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante , Traslado Adoptivo , Anciano , Alelos , Diferenciación Celular , Ensayos Clínicos como Asunto , Células Clonales/citología , Células Clonales/inmunología , Dioxigenasas/metabolismo , Epigénesis Genética , Células HEK293 , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Mutación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Transgenes
8.
J Infect Dis ; 227(6): 828-830, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36481811

RESUMEN

The recent mpox (monkeypox) outbreak has prompted genomic studies to track global spread of the disease. These studies have also revealed unexpected patterns of mutations that implicate the action of the immune defense APOBEC3 family of enzymes, which catalyze conversion of cytosine (C) to uracil (U) in DNA, in viral evolution. As poxviruses have conventionally been regarded as slow-evolving viruses, the rapid emergence of APOBEC3 mutational signatures begs a series of important and open questions regarding how host-pathogen interactions may have changed and whether these mutations are bystanders or have roles in pathogenesis.


Asunto(s)
Mpox , Virus , Humanos , Citidina Desaminasa/genética , Virus/genética , ADN , Mutación
9.
Nat Chem Biol ; 17(12): 1262-1270, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34663942

RESUMEN

DNA deaminase enzymes play key roles in immunity and have recently been harnessed for their biotechnological applications. In base editors (BEs), the combination of DNA deaminase mutator activity with CRISPR-Cas localization confers the powerful ability to directly convert one target DNA base into another. While efforts have been made to improve targeting efficiency and precision, all BEs so far use a constitutively active DNA deaminase. The absence of regulatory control over promiscuous deaminase activity remains a major limitation to accessing the widespread potential of BEs. Here, we reveal sites that permit splitting of DNA cytosine deaminases into two inactive fragments, whose reapproximation reconstitutes activity. These findings allow for the development of split-engineered BEs (seBEs), which newly enable small-molecule control over targeted mutator activity. We show that the seBE strategy facilitates robust regulated editing with BE scaffolds containing diverse deaminases, offering a generalizable solution for temporally controlling precision genome editing.


Asunto(s)
Nucleósido Desaminasas/química , Biotecnología , Sistemas CRISPR-Cas , Citosina/química , ADN/química , Roturas del ADN de Doble Cadena , Escherichia coli , Edición Génica , Conformación de Ácido Nucleico , Nucleósido Desaminasas/genética , Sirolimus/química
10.
Biochemistry ; 61(24): 2884-2896, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36473084

RESUMEN

The SOS response is a bacterial DNA damage response pathway that has been heavily implicated in bacteria's ability to evolve resistance to antibiotics. Activation of the SOS response is dependent on the interaction between two bacterial proteins, RecA and LexA. RecA acts as a DNA damage sensor by forming lengthy oligomeric filaments (RecA*) along single-stranded DNA (ssDNA) in an ATP-dependent manner. RecA* can then bind to LexA, the repressor of SOS response genes, triggering LexA degradation and leading to induction of the SOS response. Formation of the RecA*-LexA complex therefore serves as the key "SOS activation signal." Given the challenges associated with studying a complex involving multiple macromolecular interactions, the essential constituents of RecA* that allow LexA cleavage are not well defined. Here, we leverage head-to-tail linked and end-capped RecA constructs as tools to define the minimal RecA* filament that can engage LexA. In contrast to previously postulated models, we found that as few as three linked RecA units are capable of ssDNA binding, LexA binding, and LexA cleavage. We further demonstrate that RecA oligomerization alone is insufficient for LexA cleavage, with an obligate requirement for ATP and ssDNA binding to form a competent SOS activation signal with the linked constructs. Our minimal system for RecA* highlights the limitations of prior models for the SOS activation signal and offers a novel tool that can inform efforts to slow acquired antibiotic resistance by targeting the SOS response.


Asunto(s)
Proteínas Bacterianas , Respuesta SOS en Genética , Proteínas Bacterianas/química , Bacterias/metabolismo , Daño del ADN , Adenosina Trifosfato , Rec A Recombinasas/química
11.
J Biol Chem ; 296: 100087, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33199375

RESUMEN

Oxidation of 5-methylcytosine (5mC) in DNA by the ten-eleven translocation (TET) family of enzymes is indispensable for gene regulation in mammals. More recently, evidence has emerged to support a biological function for TET-mediated m5C oxidation in messenger RNA. Here, we describe a previously uncharacterized role of TET-mediated m5C oxidation in transfer RNA (tRNA). We found that the TET-mediated oxidation product 5-hydroxylmethylcytosine (hm5C) is specifically enriched in tRNA inside cells and that the oxidation activity of TET2 on m5C in tRNAs can be readily observed in vitro. We further observed that hm5C levels in tRNA were significantly decreased in Tet2 KO mouse embryonic stem cells (mESCs) in comparison with wild-type mESCs. Reciprocally, induced expression of the catalytic domain of TET2 led to an obvious increase in hm5C and a decrease in m5C in tRNAs relative to uninduced cells. Strikingly, we also show that TET2-mediated m5C oxidation in tRNA promotes translation in vitro. These results suggest TET2 may influence translation through impacting tRNA methylation and reveal an unexpected role for TET enzymes in regulating multiple nodes of the central dogma.


Asunto(s)
5-Metilcitosina/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN de Transferencia/metabolismo , 5-Metilcitosina/química , Animales , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Células Madre Embrionarias/metabolismo , Ratones , Ratones Noqueados , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas/química , ARN de Transferencia/química
12.
Bioorg Med Chem Lett ; 65: 128702, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35351585

RESUMEN

Resistant and tolerant bacterial infections lead to billions in healthcare costs and cause hundreds of thousands of deaths each year. The bulk of current antibiotic research efforts focus on molecules which, although novel, are not immune from acquired resistance and seldomly affect tolerant populations. The bacterial SOS response has been implicated in several resistance and tolerance mechanisms, making it an attractive antibiotic target. Using small molecule inhibitors targeting a key step in the deployment of the SOS response, our approach focused on preventing the deployment of mechanisms such as biofilm formation, horizontal gene transfer, and error-prone DNA repair. Herein we report the synthesis and testing of analogs of a triazole-containing tricyclic inhibitor of LexA proteolysis, the key event in the SOS response. Our results hint that our inhibitor's may function by adopting a ß-hairpin conformation, reminiscent of the native cleavage loop of LexA.


Asunto(s)
Péptido Hidrolasas , Respuesta SOS en Genética , Antibacterianos/farmacología , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Serina Endopeptidasas/metabolismo
13.
PLoS Genet ; 14(6): e1007405, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29856734

RESUMEN

Biochemical pathways are often genetically encoded as simple transcription regulation networks, where one transcription factor regulates the expression of multiple genes in a pathway. The relative timing of each promoter's activation and shut-off within the network can impact physiology. In the DNA damage repair pathway (known as the SOS response) of Escherichia coli, approximately 40 genes are regulated by the LexA repressor. After a DNA damaging event, LexA degradation triggers SOS gene transcription, which is temporally separated into subsets of 'early', 'middle', and 'late' genes. Although this feature plays an important role in regulating the SOS response, both the range of this separation and its underlying mechanism are not experimentally defined. Here we show that, at low doses of DNA damage, the timing of promoter activities is not separated. Instead, timing differences only emerge at higher levels of DNA damage and increase as a function of DNA damage dose. To understand mechanism, we derived a series of synthetic SOS gene promoters which vary in LexA-operator binding kinetics, but are otherwise identical, and then studied their activity over a large dose-range of DNA damage. In distinction to established models based on rapid equilibrium assumptions, the data best fit a kinetic model of repressor occupancy at promoters, where the drop in cellular LexA levels associated with higher doses of DNA damage leads to non-equilibrium binding kinetics of LexA at operators. Operators with slow LexA binding kinetics achieve their minimal occupancy state at later times than operators with fast binding kinetics, resulting in a time separation of peak promoter activity between genes. These data provide insight into this remarkable feature of the SOS pathway by demonstrating how a single transcription factor can be employed to control the relative timing of each gene's transcription as a function of stimulus dose.


Asunto(s)
Proteínas Bacterianas/metabolismo , Daño del ADN/genética , Escherichia coli/genética , Proteínas Represoras/metabolismo , Respuesta SOS en Genética/genética , Serina Endopeptidasas/metabolismo , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Redes Reguladoras de Genes/genética , Genes Bacterianos , Cinética , Modelos Genéticos , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Serina Endopeptidasas/genética , Factores de Tiempo
14.
Angew Chem Int Ed Engl ; 59(28): 11312-11315, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32271979

RESUMEN

TET family enzymes are known for oxidation of the 5-methyl substituent on 5-methylcytosine (5mC) in DNA. 5mC oxidation generates the stable base 5-hydroxymethylcytosine (5hmC), starting an indirect, multi-step process that ends with reversion of 5mC to unmodified cytosine. While probing the nucleobase determinants of 5mC recognition, we discovered that TET enzymes are also proficient as direct N-demethylases of cytosine bases. We find that N-demethylase activity can be readily observed on substrates lacking a 5-methyl group and, remarkably, TET enzymes can be similarly proficient in either oxidation of 5mC or demethylation of N4-methyl substituents. Our results indicate that TET enzymes can act as both direct and indirect demethylases, highlight the active-site plasticity of these FeII /α-ketoglutarate-dependent dioxygenases, and suggest activity on unexplored substrates that could reveal new TET biology.


Asunto(s)
ADN/metabolismo , Dioxigenasas/metabolismo , Nitrógeno/metabolismo , 5-Metilcitosina/metabolismo , Dominio Catalítico , Metilación de ADN , Desmetilación , Humanos , Oxidación-Reducción , Análisis Espectral/métodos
15.
Biochemistry ; 58(5): 411-421, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30387995

RESUMEN

Enzymes of the ten-eleven translocation (TET) family add diversity to the repertoire of nucleobase modifications by catalyzing the oxidation of 5-methylcytosine (5mC). TET enzymes were initially found to oxidize 5-methyl-2'-deoxycytidine in genomic DNA, yielding products that contribute to epigenetic regulation in mammalian cells, but have since been found to also oxidize 5-methylcytidine in RNA. Considering the different configurations of single-stranded (ss) and double-stranded (ds) DNA and RNA that coexist in a cell, defining the scope of TET's preferred activity and the mechanisms of substrate selectivity is critical to better understand the enzymes' biological functions. To this end, we have systematically examined the activity of human TET2 on DNA, RNA, and hybrid substrates in vitro. We found that, while ssDNA and ssRNA are well tolerated, TET2 is most proficient at dsDNA oxidation and discriminates strongly against dsRNA. Chimeric and hybrid substrates containing mixed DNA and RNA character helped reveal two main features by which the enzyme discriminates between substrates. First, the identity of the target nucleotide alone is the strongest reactivity determinant, with a preference for 5-methyldeoxycytidine, while both DNA or RNA are relatively tolerated on the rest of the target strand. Second, while a complementary strand is not required for activity, DNA is the preferred partner, and complementary RNA diminishes reactivity. Our biochemical analysis, complemented by molecular dynamics simulations, provides support for an active site optimally configured for dsDNA reactivity but permissive for various nucleic acid configurations, suggesting a broad range of plausible roles for TET-mediated 5mC oxidation in cells.


Asunto(s)
5-Metilcitosina/química , Proteínas de Unión al ADN/metabolismo , ADN/química , ADN/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN/química , ARN/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Dioxigenasas , Epigénesis Genética , Humanos , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Especificidad por Sustrato
16.
Infect Immun ; 87(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30510108

RESUMEN

Bacteria have a remarkable ability to survive, persist, and ultimately adapt to environmental challenges. A ubiquitous environmental hazard is DNA damage, and most bacteria have evolved a network of genes to combat genotoxic stress. This network is known as the SOS response and aids in bacterial survival by regulating genes involved in DNA repair and damage tolerance. Recently, the SOS response has been shown to play an important role in bacterial pathogenesis, and yet the role of the SOS response in nonpathogenic organisms and in physiological settings remains underexplored. Using a commensal Escherichia coli strain, MP1, we showed that the SOS response plays a vital role during colonization of the murine gut. In an unperturbed environment, the SOS-off mutant is impaired for stable colonization relative to a wild-type strain, suggesting the presence of genotoxic stress in the mouse gut. We evaluated the possible origins of genotoxic stress in the mouse gut by examining factors associated with the host versus the competing commensal organisms. In a dextran sulfate sodium (DSS) colitis model, the SOS-off colonization defect persisted but was not exacerbated. In contrast, in a germ-free model, the SOS-off mutant colonized with efficiency equal to that seen with the wild-type strain, suggesting that competing commensal organisms might be a significant source of genotoxic stress. This report extends our understanding of the importance of a functional SOS response for bacterial fitness in the context of a complex physiological environment and highlights the SOS response as a possible mechanism that contributes to ongoing genomic changes, including potential antibiotic resistance, in the microbiome of healthy hosts.


Asunto(s)
Daño del ADN/fisiología , Escherichia coli/patogenicidad , Tracto Gastrointestinal/microbiología , Respuesta SOS en Genética/fisiología , Animales , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica , Ratones , Ratones Endogámicos C57BL
17.
Nat Chem Biol ; 13(2): 181-187, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27918559

RESUMEN

Ten-eleven translocation (TET) enzymes catalyze stepwise oxidation of 5-methylcytosine (mC) to yield 5-hydroxymethylcytosine (hmC) and the rarer bases 5-formylcytosine (fC) and 5-carboxylcytosine (caC). Stepwise oxidation obscures how each individual base forms and functions in epigenetic regulation, and prompts the question of whether TET enzymes primarily serve to generate hmC or are adapted to produce fC and caC as well. By mutating a single, conserved active site residue in human TET2, Thr1372, we uncovered enzyme variants that permit oxidation to hmC but largely eliminate fC and caC. Biochemical analyses, combined with molecular dynamics simulations, elucidated an active site scaffold that is required for wild-type (WT) stepwise oxidation and that, when perturbed, explains the mutants' hmC-stalling phenotype. Our results suggest that the TET2 active site is shaped to enable higher-order oxidation and provide the first TET variants that could be used to probe the biological functions of hmC separately from fC and caC.


Asunto(s)
5-Metilcitosina/análogos & derivados , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Mutación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/química , 5-Metilcitosina/metabolismo , Dominio Catalítico/genética , Proteínas de Unión al ADN/química , Dioxigenasas , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Oxidación-Reducción , Proteínas Proto-Oncogénicas/química
18.
Nature ; 502(7472): 472-9, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24153300

RESUMEN

DNA methylation has a profound impact on genome stability, transcription and development. Although enzymes that catalyse DNA methylation have been well characterized, those that are involved in methyl group removal have remained elusive, until recently. The transformative discovery that ten-eleven translocation (TET) family enzymes can oxidize 5-methylcytosine has greatly advanced our understanding of DNA demethylation. 5-Hydroxymethylcytosine is a key nexus in demethylation that can either be passively depleted through DNA replication or actively reverted to cytosine through iterative oxidation and thymine DNA glycosylase (TDG)-mediated base excision repair. Methylation, oxidation and repair now offer a model for a complete cycle of dynamic cytosine modification, with mounting evidence for its significance in the biological processes known to involve active demethylation.


Asunto(s)
5-Metilcitosina/metabolismo , Metilación de ADN , Timina ADN Glicosilasa/metabolismo , 5-Metilcitosina/química , Animales , Blastocisto/citología , Blastocisto/metabolismo , Reprogramación Celular , Citosina/análogos & derivados , Citosina/química , Citosina/metabolismo , Reparación del ADN , Replicación del ADN , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Oxidación-Reducción
19.
Nucleic Acids Res ; 45(13): 7655-7665, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28472485

RESUMEN

AID/APOBEC family enzymes are best known for deaminating cytosine bases to uracil in single-stranded DNA, with characteristic sequence preferences that can produce mutational signatures in targets such as retroviral and cancer cell genomes. These deaminases have also been proposed to function in DNA demethylation via deamination of either 5-methylcytosine (mC) or TET-oxidized mC bases (ox-mCs), which include 5-hydroxymethylcytosine, 5-formylcytosine and 5-carboxylcytosine. One specific family member, APOBEC3A (A3A), has been shown to readily deaminate mC, raising the prospect of broader activity on ox-mCs. To investigate this claim, we developed a novel assay that allows for parallel profiling of activity on all modified cytosines. Our steady-state kinetic analysis reveals that A3A discriminates against all ox-mCs by >3700-fold, arguing that ox-mC deamination does not contribute substantially to demethylation. A3A is, by contrast, highly proficient at C/mC deamination. Under conditions of excess enzyme, C/mC bases can be deaminated to completion in long DNA segments, regardless of sequence context. Interestingly, under limiting A3A, the sequence preferences observed with targeting unmodified cytosine are further exaggerated when deaminating mC. Our study informs how methylation, oxidation, and deamination can interplay in the genome and suggests A3A's potential utility as a biotechnological tool to discriminate between cytosine modification states.


Asunto(s)
Citidina Desaminasa/metabolismo , Citosina/química , ADN/química , ADN/metabolismo , Proteínas/metabolismo , 5-Metilcitosina/química , Secuencia de Bases , Metilación de ADN , Humanos , Técnicas In Vitro , Cinética , Modelos Biológicos , Oxidación-Reducción , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
20.
J Am Chem Soc ; 140(50): 17329-17332, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30518204

RESUMEN

Ten-eleven translocation (TET) enzymes catalyze repeated oxidations of 5-methylcytosine in genomic DNA. Because of the challenges of tracking reactivity within a complex DNA substrate, chemical tools to probe TET activity are limited, despite these enzyme's crucial role in epigenetic regulation. Here, building on precedents from related Fe(II)/α-ketoglutarate-dependent dioxygenases, we show that TET enzymes can promiscuously act upon cytosine bases with unnatural 5-position modifications. Oxidation of 5-vinylcytosine (vC) in DNA results in the predominant formation of a 5-formylmethylcytosine product that can be efficiently labeled to provide an end-point read-out for TET activity. The reaction with 5-ethynylcytosine (eyC), moreover, results in the formation of a high-energy ketene intermediate that can selectively trap any active TET isoform as a covalent enzyme-DNA complex, even in the complex milieu of a total cell lysate. Exploiting substrate promiscuity therefore offers a new and needed means to directly track TET activity in vitro or in vivo.


Asunto(s)
Sondas de ADN/química , ADN/química , Dioxigenasas/química , Reactivos de Enlaces Cruzados/química , Citosina/análogos & derivados , Citosina/química , Células HEK293 , Humanos , Oxidación-Reducción , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA