Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(5): 1127-1142.e19, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30078706

RESUMEN

Replication origins, fragile sites, and rDNA have been implicated as sources of chromosomal instability. However, the defining genomic features of replication origins and fragile sites are among the least understood elements of eukaryote genomes. Here, we map sites of replication initiation and breakage in primary cells at high resolution. We find that replication initiates between transcribed genes within nucleosome-depleted structures established by long asymmetrical poly(dA:dT) tracts flanking the initiation site. Paradoxically, long (>20 bp) (dA:dT) tracts are also preferential sites of polar replication fork stalling and collapse within early-replicating fragile sites (ERFSs) and late-replicating common fragile sites (CFSs) and at the rDNA replication fork barrier. Poly(dA:dT) sequences are fragile because long single-strand poly(dA) stretches at the replication fork are unprotected by the replication protein A (RPA). We propose that the evolutionary expansion of poly(dA:dT) tracts in eukaryotic genomes promotes replication initiation, but at the cost of chromosome fragility.


Asunto(s)
Replicación del ADN , ADN Ribosómico/química , Nucleosomas/metabolismo , Poli dA-dT/química , Origen de Réplica , Secuencias de Aminoácidos , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Inestabilidad Cromosómica , Sitios Frágiles del Cromosoma , Fragilidad Cromosómica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Saccharomyces cerevisiae , Schizosaccharomyces , Sitio de Iniciación de la Transcripción , Transcripción Genética
2.
Cell ; 173(5): 1165-1178.e20, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29706548

RESUMEN

Cohesin extrusion is thought to play a central role in establishing the architecture of mammalian genomes. However, extrusion has not been visualized in vivo, and thus, its functional impact and energetics are unknown. Using ultra-deep Hi-C, we show that loop domains form by a process that requires cohesin ATPases. Once formed, however, loops and compartments are maintained for hours without energy input. Strikingly, without ATP, we observe the emergence of hundreds of CTCF-independent loops that link regulatory DNA. We also identify architectural "stripes," where a loop anchor interacts with entire domains at high frequency. Stripes often tether super-enhancers to cognate promoters, and in B cells, they facilitate Igh transcription and recombination. Stripe anchors represent major hotspots for topoisomerase-mediated lesions, which promote chromosomal translocations and cancer. In plasmacytomas, stripes can deregulate Igh-translocated oncogenes. We propose that higher organisms have coopted cohesin extrusion to enhance transcription and recombination, with implications for tumor development.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Genoma , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Cromosomas/metabolismo , Proteínas de Unión al ADN , Humanos , Ratones , Mutagénesis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Cohesinas
3.
Cell ; 170(3): 507-521.e18, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28735753

RESUMEN

In this study, we show that evolutionarily conserved chromosome loop anchors bound by CCCTC-binding factor (CTCF) and cohesin are vulnerable to DNA double strand breaks (DSBs) mediated by topoisomerase 2B (TOP2B). Polymorphisms in the genome that redistribute CTCF/cohesin occupancy rewire DNA cleavage sites to novel loop anchors. While transcription- and replication-coupled genomic rearrangements have been well documented, we demonstrate that DSBs formed at loop anchors are largely transcription-, replication-, and cell-type-independent. DSBs are continuously formed throughout interphase, are enriched on both sides of strong topological domain borders, and frequently occur at breakpoint clusters commonly translocated in cancer. Thus, loop anchors serve as fragile sites that generate DSBs and chromosomal rearrangements. VIDEO ABSTRACT.


Asunto(s)
Fragilidad Cromosómica , Roturas del ADN de Doble Cadena , Neoplasias/genética , Animales , Linfocitos B/metabolismo , Factor de Unión a CCCTC , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Represoras/metabolismo
4.
Cell ; 162(4): 751-65, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26234156

RESUMEN

The RAG1 endonuclease, together with its cofactor RAG2, is essential for V(D)J recombination but is a potent threat to genome stability. The sources of RAG1 mis-targeting and the mechanisms that have evolved to suppress it are poorly understood. Here, we report that RAG1 associates with chromatin at thousands of active promoters and enhancers in the genome of developing lymphocytes. The mouse and human genomes appear to have responded by reducing the abundance of "cryptic" recombination signals near RAG1 binding sites. This depletion operates specifically on the RSS heptamer, whereas nonamers are enriched at RAG1 binding sites. Reversing this RAG-driven depletion of cleavage sites by insertion of strong recombination signals creates an ectopic hub of RAG-mediated V(D)J recombination and chromosomal translocations. Our findings delineate rules governing RAG binding in the genome, identify areas at risk of RAG-mediated damage, and highlight the evolutionary struggle to accommodate programmed DNA damage in developing lymphocytes.


Asunto(s)
Inestabilidad Genómica , Proteínas de Homeodominio/metabolismo , Linfocitos/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Secuencia de Bases , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN/metabolismo , Humanos , Linfocitos/citología , Ratones , Datos de Secuencia Molecular , Translocación Genética , Recombinación V(D)J
6.
Mol Cell ; 77(1): 26-38.e7, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31653568

RESUMEN

53BP1 activity drives genome instability and lethality in BRCA1-deficient mice by inhibiting homologous recombination (HR). The anti-recombinogenic functions of 53BP1 require phosphorylation-dependent interactions with PTIP and RIF1/shieldin effector complexes. While RIF1/shieldin blocks 5'-3' nucleolytic processing of DNA ends, it remains unclear how PTIP antagonizes HR. Here, we show that mutation of the PTIP interaction site in 53BP1 (S25A) allows sufficient DNA2-dependent end resection to rescue the lethality of BRCA1Δ11 mice, despite increasing RIF1 "end-blocking" at DNA damage sites. However, double-mutant cells fail to complete HR, as excessive shieldin activity also inhibits RNF168-mediated loading of PALB2/RAD51. As a result, BRCA1Δ1153BP1S25A mice exhibit hallmark features of HR insufficiency, including premature aging and hypersensitivity to PARPi. Disruption of shieldin or forced targeting of PALB2 to ssDNA in BRCA1D1153BP1S25A cells restores RNF168 recruitment, RAD51 nucleofilament formation, and PARPi resistance. Our study therefore reveals a critical function of shieldin post-resection that limits the loading of RAD51.


Asunto(s)
Recombinación Homóloga/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Proteína BRCA1/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Recombinación Homóloga/efectos de los fármacos , Ratones , Mutación/efectos de los fármacos , Mutación/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Recombinasa Rad51/genética , Ubiquitina-Proteína Ligasas/genética
7.
Mol Cell ; 75(2): 252-266.e8, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31202577

RESUMEN

Topoisomerase II (TOP2) relieves torsional stress by forming transient cleavage complex intermediates (TOP2ccs) that contain TOP2-linked DNA breaks (DSBs). While TOP2ccs are normally reversible, they can be "trapped" by chemotherapeutic drugs such as etoposide and subsequently converted into irreversible TOP2-linked DSBs. Here, we have quantified etoposide-induced trapping of TOP2ccs, their conversion into irreversible TOP2-linked DSBs, and their processing during DNA repair genome-wide, as a function of time. We find that while TOP2 chromatin localization and trapping is independent of transcription, it requires pre-existing binding of cohesin to DNA. In contrast, the conversion of trapped TOP2ccs to irreversible DSBs during DNA repair is accelerated 2-fold at transcribed loci relative to non-transcribed loci. This conversion is dependent on proteasomal degradation and TDP2 phosphodiesterase activity. Quantitative modeling shows that only two features of pre-existing chromatin structure-namely, cohesin binding and transcriptional activity-can be used to predict the kinetics of TOP2-induced DSBs.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN-Topoisomerasas de Tipo II/química , ADN/genética , Complejos Multiproteicos/química , Proteínas de Unión a Poli-ADP-Ribosa/química , Rotura Cromosómica , Cromosomas/genética , ADN/química , Reparación del ADN/genética , ADN-Topoisomerasas de Tipo II/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Etopósido/química , Conversión Génica/genética , Células HCT116 , Humanos , Cinética , Complejos Multiproteicos/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/farmacología , Torsión Mecánica , Transcripción Genética , Translocación Genética/genética
8.
J Immunol ; 208(1): 143-154, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34862258

RESUMEN

Somatic hypermutation (SHM) drives the genetic diversity of Ig genes in activated B cells and supports the generation of Abs with increased affinity for Ag. SHM is targeted to Ig genes by their enhancers (diversification activators [DIVACs]), but how the enhancers mediate this activity is unknown. We show using chicken DT40 B cells that highly active DIVACs increase the phosphorylation of RNA polymerase II (Pol II) and Pol II occupancy in the mutating gene with little or no accompanying increase in elongation-competent Pol II or production of full-length transcripts, indicating accumulation of stalled Pol II. DIVAC has similar effect also in human Ramos Burkitt lymphoma cells. The DIVAC-induced stalling is weakly associated with an increase in the detection of ssDNA bubbles in the mutating target gene. We did not find evidence for antisense transcription, or that DIVAC functions by altering levels of H3K27ac or the histone variant H3.3 in the mutating gene. These findings argue for a connection between Pol II stalling and cis-acting targeting elements in the context of SHM and thus define a mechanistic basis for locus-specific targeting of SHM in the genome. Our results suggest that DIVAC elements render the target gene a suitable platform for AID-mediated mutation without a requirement for increasing transcriptional output.


Asunto(s)
Proteínas Aviares/metabolismo , Subgrupos de Linfocitos B/inmunología , Linfoma de Burkitt/inmunología , Elementos de Facilitación Genéticos/genética , Inmunoglobulinas/metabolismo , ARN Polimerasa II/metabolismo , Animales , Diversidad de Anticuerpos , Proteínas Aviares/genética , Linfoma de Burkitt/genética , Pollos , Citidina Desaminasa/genética , Humanos , Inmunoglobulinas/genética , Activación de Linfocitos , Mutagénesis Sitio-Dirigida , Mutación/genética , ARN Polimerasa II/genética , Hipermutación Somática de Inmunoglobulina , Transcripción Genética
9.
Nucleic Acids Res ; 44(20): 9624-9637, 2016 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-27436288

RESUMEN

The RAG1/RAG2 endonuclease initiates V(D)J recombination at antigen receptor loci but also binds to thousands of places outside of these loci. RAG2 localizes directly to lysine 4 trimethylated histone 3 (H3K4me3) through a plant homeodomain (PHD) finger. The relative contribution of RAG2-dependent and RAG1-intrinsic mechanisms in determining RAG1 binding patterns is not known. Through analysis of deep RAG1 ChIP-seq data, we provide a quantitative description of the forces underlying genome-wide targeting of RAG1. Surprisingly, sequence-specific DNA binding contributes minimally to RAG1 targeting outside of antigen receptor loci. Instead, RAG1 binding is driven by two distinct modes of interaction with chromatin: the first is driven by H3K4me3, promoter-focused and dependent on the RAG2 PHD, and the second is defined by H3K27Ac, enhancer-focused and dependent on 'non-core' portions of RAG1. Based on this and additional chromatin and genomic features, we formulated a predictive model of RAG1 targeting to the genome. RAG1 binding sites predicted by our model correlate well with observed patterns of RAG1-mediated breaks in human pro-B acute lymphoblastic leukemia. Overall, this study provides an integrative model for RAG1 genome-wide binding and off-target activity and reveals a novel role for the RAG1 non-core region in RAG1 targeting.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Genoma , Proteínas de Homeodominio/metabolismo , Animales , Sitios de Unión , Inmunoprecipitación de Cromatina , Inestabilidad Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Histonas/metabolismo , Proteínas de Homeodominio/química , Humanos , Ratones , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Recombinación Genética , Recombinación V(D)J
10.
Nucleic Acids Res ; 44(5): e46, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26586802

RESUMEN

Incremental selection within a population, defined as limited fitness changes following mutation, is an important aspect of many evolutionary processes. Strongly advantageous or deleterious mutations are detected using the synonymous to non-synonymous mutations ratio. However, there are currently no precise methods to estimate incremental selection. We here provide for the first time such a detailed method and show its precision in multiple cases of micro-evolution. The proposed method is a novel mixed lineage tree/sequence based method to detect within population selection as defined by the effect of mutations on the average number of offspring. Specifically, we propose to measure the log of the ratio between the number of leaves in lineage trees branches following synonymous and non-synonymous mutations. The method requires a high enough number of sequences, and a large enough number of independent mutations. It assumes that all mutations are independent events. It does not require of a baseline model and is practically not affected by sampling biases. We show the method's wide applicability by testing it on multiple cases of micro-evolution. We show that it can detect genes and inter-genic regions using the selection rate and detect selection pressures in viral proteins and in the immune response to pathogens.


Asunto(s)
Algoritmos , Evolución Biológica , Modelos Genéticos , Linaje , Selección Genética , Alphapapillomavirus/clasificación , Alphapapillomavirus/genética , Animales , Secuencia de Bases , Simulación por Computador , Epítopos/química , Epítopos/genética , VIH/clasificación , VIH/genética , Virus de la Hepatitis B/clasificación , Virus de la Hepatitis B/genética , Humanos , Inmunoglobulinas/clasificación , Inmunoglobulinas/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Filogenia , ARN Viral/química , ARN Viral/genética , Receptores de Antígenos de Linfocitos B/clasificación , Receptores de Antígenos de Linfocitos B/genética , Alineación de Secuencia
11.
Immunogenetics ; 67(1): 15-24, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25376343

RESUMEN

Viral epitopes have a distinct codon usage that reflects their dual role in infection and immunity. On the one hand, epitopes are part of proteins important to viral function; on the other hand, they are targets of the immune response. Studies of selection are most commonly based on changes of amino acid and seen through the accumulation of non-synonymous mutations. An independent measure of selection is the codon usage and underlying changeability of the nucleotide sequences. We here use multiple tools and a large-scale analysis of viral genomes to demonstrate that viral epitopes have a distinct codon usage and that this codon usage reflects distinct short- and long-term types of selection during viral evolution. We show that CD8(+) T cell epitopes are encoded by codons more distant from stop codons and more changeable than codons outside epitopes. This biased codon usage reflects the viral population toggling back and forth from a wild-type sequence to an escape mode, which enable them to avoid immune detection when needed, and go back to the functionally favorable form when the threat is removed (i.e., in a new host).


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/genética , Evolución Molecular , Selección Genética , Codón/genética , Codón/inmunología , Biología Computacional , Epítopos de Linfocito T/inmunología , Humanos , Nucleótidos/genética , Nucleótidos/inmunología , Filogenia , Virus/genética , Virus/inmunología
12.
Cell Mol Gastroenterol Hepatol ; 16(1): 63-81, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36965814

RESUMEN

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is a model of a diverse spectrum of cancers because it is induced by well-known etiologies, mainly hepatitis C virus (HCV) and hepatitis B virus. Here, we aimed to identify HCV-specific mutational signatures and explored the link between the HCV-related regional variation in mutations rates and HCV-induced alterations in genome-wide chromatin organization. METHODS: To identify an HCV-specific mutational signature in HCC, we performed high-resolution targeted sequencing to detect passenger mutations on 64 HCC samples from 3 etiology groups: hepatitis B virus, HCV, or other. To explore the link between the genomic signature and genome-wide chromatin organization we performed chromatin immunoprecipitation sequencing for the transcriptionally permissive H3K4Me3, H3K9Ac, and suppressive H3K9Me3 modifications after HCV infection. RESULTS: Regional variation in mutation rate analysis showed significant etiology-dependent regional mutation rates in 12 genes: LRP2, KRT84, TMEM132B, DOCK2, DMD, INADL, JAK2, DNAH6, MTMR9, ATM, SLX4, and ARSD. We found an enrichment of C->T transversion mutations in the HCV-associated HCC cases. Furthermore, these cases showed regional variation in mutation rates associated with genomic intervals in which HCV infection dictated epigenetic alterations. This signature may be related to the HCV-induced decreased expression of genes encoding key enzymes in the base excision repair pathway. CONCLUSIONS: We identified novel distinct HCV etiology-dependent mutation signatures in HCC associated with HCV-induced alterations in histone modification. This study presents a link between cancer-causing mutagenesis and the increased predisposition to liver cancer in chronic HCV-infected individuals, and unveils novel etiology-specific mechanisms leading to HCC and cancer in general.


Asunto(s)
Carcinoma Hepatocelular , Hepatitis C , Neoplasias Hepáticas , Humanos , Neoplasias Hepáticas/patología , Carcinoma Hepatocelular/patología , Hepatitis C/complicaciones , Hepatitis C/genética , Mutación/genética , Hepacivirus/genética , Virus de la Hepatitis B/genética , Epigénesis Genética/genética , Cromatina , Genómica , Proteínas Tirosina Fosfatasas no Receptoras/genética , Queratinas Tipo II/genética , Queratinas Específicas del Pelo/genética
13.
J Virol ; 85(9): 4558-66, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307195

RESUMEN

Viruses employ various means to evade immune detection. Reduction of CD8(+) T cell epitopes is one of the common strategies used for this purpose. Hepatitis B virus (HBV), a member of the Hepadnaviridae family, has four open reading frames, with about 50% overlap between the genes they encode. We computed the CD8(+) T cell epitope density within HBV proteins and the mutations within the epitopes. Our results suggest that HBV accumulates escape mutations that reduce the number of epitopes. These mutations are not equally distributed among genes and reading frames. While the highly expressed core and X proteins are selected to have low epitope density, polymerase, which is expressed at low levels, does not undergo the same selection. In overlapping regions, mutations in one protein-coding sequence also affect the other protein-coding sequence. We show that mutations lead to the removal of epitopes in X and surface proteins even at the expense of the addition of epitopes in polymerase. The total escape mutation rate for overlapping regions is lower than that for nonoverlapping regions. The lower epitope replacement rate for overlapping regions slows the evolutionary escape rate of these regions but leads to the accumulation of mutations more robust in the transfer between hosts, such as mutations preventing proteasomal cleavage into epitopes.


Asunto(s)
Evolución Molecular , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Selección Genética , Proteínas Virales/genética , Proteínas Virales/inmunología , Linfocitos T CD8-positivos/inmunología , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/inmunología , Epítopos/genética , Epítopos/inmunología , Mutación , Sistemas de Lectura Abierta , Transactivadores/genética , Transactivadores/inmunología , Proteínas Reguladoras y Accesorias Virales
14.
PLoS Comput Biol ; 7(10): e1002220, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22022257

RESUMEN

The main adaptive immune response to bacteria is mediated by B cells and CD4+ T-cells. However, some bacterial proteins reach the cytosol of host cells and are exposed to the host CD8+ T-cells response. Both gram-negative and gram-positive bacteria can translocate proteins to the cytosol through type III and IV secretion and ESX-1 systems, respectively. The translocated proteins are often essential for the bacterium survival. Once injected, these proteins can be degraded and presented on MHC-I molecules to CD8+ T-cells. The CD8+ T-cells, in turn, can induce cell death and destroy the bacteria's habitat. In viruses, escape mutations arise to avoid this detection. The accumulation of escape mutations in bacteria has never been systematically studied. We show for the first time that such mutations are systematically present in most bacteria tested. We combine multiple bioinformatic algorithms to compute CD8+ T-cell epitope libraries of bacteria with secretion systems that translocate proteins to the host cytosol. In all bacteria tested, proteins not translocated to the cytosol show no escape mutations in their CD8+ T-cell epitopes. However, proteins translocated to the cytosol show clear escape mutations and have low epitope densities for most tested HLA alleles. The low epitope densities suggest that bacteria, like viruses, are evolutionarily selected to ensure their survival in the presence of CD8+ T-cells. In contrast with most other translocated proteins examined, Pseudomonas aeruginosa's ExoU, which ultimately induces host cell death, was found to have high epitope density. This finding suggests a novel mechanism for the manipulation of CD8+ T-cells by pathogens. The ExoU effector may have evolved to maintain high epitope density enabling it to efficiently induce CD8+ T-cell mediated cell death. These results were tested using multiple epitope prediction algorithms, and were found to be consistent for most proteins tested.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Linfocitos T CD8-positivos/inmunología , Citosol/inmunología , Epítopos/clasificación , Algoritmos , Alelos , Epítopos/inmunología , Antígenos HLA/genética , Antígenos HLA/inmunología , Humanos , Mutación
15.
Microbiol Spectr ; 10(6): e0115022, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36314945

RESUMEN

Recent studies suggest the enhancement of liver injury in COVID-19 patients infected with Hepatitis C virus (HCV). Hepatocytes express low levels of angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 entry receptor, raising the possibility of HCV-SARS-CoV-2 coinfection in the liver. This work aimed to explore whether HCV and SARS-CoV-2 coinfect hepatocytes and the interplay between these viruses. We demonstrate that SARS-CoV-2 coinfects HCV-infected Huh7.5 (Huh7.5HCV) cells. Both viruses replicated efficiently in the coinfected cells, with HCV replication enhanced in coinfected compared to HCV-mono-infected cells. Strikingly, Huh7.5HCV cells were eight fold more susceptible to SARS-CoV-2 pseudoviruses than naive Huh7.5 cells, suggesting enhanced SARS-CoV-2 entry into HCV-preinfected hepatocytes. In addition, we observed increased binding of spike receptor-binding domain (RBD) protein to Huh7.5HCV cells, as well as enhanced cell-to-cell fusion of Huh7.5HCV cells with spike-expressing Huh7.5 cells. We explored the mechanism of enhanced SARS-CoV-2 entry and identified an increased ACE2 mRNA and protein levels in Huh7.5HCV cells, primary hepatocytes, and in data from infected liver biopsies obtained from database. Importantly, higher expression of ACE2 increased HCV infection by enhancing its binding to the host cell, underscoring its role in the HCV life cycle as well. Transcriptome analysis revealed that shared host signaling pathways were induced in HCV-SARS-CoV-2 coinfection. This study revealed complex interactions between HCV and SARS-CoV-2 infections in hepatocytes, which may lead to the increased liver damage recently reported in HCV-positive COVID-19 patients. IMPORTANCE Here, we provide the first experimental evidence for the coexistence of SARS-CoV-2 infection with HCV, and the interplay between them. The study revealed a complex relationship of enhancement between the two viruses, where HCV infection increased the expression of the SARS-CoV-2 entry receptor ACE2, thus facilitating SARS-CoV-2 entry, and potentially, also HCV entry. Thereafter, SARS-CoV-2 infection enhanced HCV replication in hepatocytes. This study may explain the aggravation of liver damage that was recently reported in COVID-19 patients with HCV coinfection and suggests preinfection with HCV as a risk factor for severe COVID-19. Moreover, it highlights the possible importance of HCV treatment for coinfected patients. In a broader view, these findings emphasize the importance of identifying coinfecting pathogens that increase the risk of SARS-CoV-2 infection and that may accelerate COVID-19-related co-morbidities.


Asunto(s)
COVID-19 , Coinfección , Hepatitis C , Humanos , SARS-CoV-2/metabolismo , Hepacivirus , Enzima Convertidora de Angiotensina 2/metabolismo , Receptores Virales/genética , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Hepatitis C/complicaciones , Hepatocitos , Unión Proteica
16.
J Cell Biol ; 220(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33464298

RESUMEN

Genome-wide CRISPR screens have transformed our ability to systematically interrogate human gene function, but are currently limited to a subset of cellular phenotypes. We report a novel pooled screening approach for a wider range of cellular and subtle subcellular phenotypes. Machine learning and convolutional neural network models are trained on the subcellular phenotype to be queried. Genome-wide screening then utilizes cells stably expressing dCas9-KRAB (CRISPRi), photoactivatable fluorescent protein (PA-mCherry), and a lentiviral guide RNA (gRNA) pool. Cells are screened by using microscopy and classified by artificial intelligence (AI) algorithms, which precisely identify the genetically altered phenotype. Cells with the phenotype of interest are photoactivated and isolated via flow cytometry, and the gRNAs are identified by sequencing. A proof-of-concept screen accurately identified PINK1 as essential for Parkin recruitment to mitochondria. A genome-wide screen identified factors mediating TFEB relocation from the nucleus to the cytosol upon prolonged starvation. Twenty-one of the 64 hits called by the neural network model were independently validated, revealing new effectors of TFEB subcellular localization. This approach, AI-photoswitchable screening (AI-PS), offers a novel screening platform capable of classifying a broad range of mammalian subcellular morphologies, an approach largely unattainable with current methodologies at genome-wide scale.


Asunto(s)
Sistemas CRISPR-Cas/genética , Pruebas Genéticas , Genoma , Imagenología Tridimensional , Inteligencia Artificial , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/metabolismo , Aprendizaje Profundo , Proteínas Fluorescentes Verdes , Células HEK293 , Humanos , Modelos Biológicos , Redes Neurales de la Computación , Fenotipo , Reproducibilidad de los Resultados , Análisis de la Célula Individual , Máquina de Vectores de Soporte , Ubiquitina-Proteína Ligasas/metabolismo , ARN Guía de Sistemas CRISPR-Cas
17.
Nat Commun ; 11(1): 857, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-32051414

RESUMEN

Meiotic recombination is initiated by SPO11-induced double-strand breaks (DSBs). In most mammals, the methyltransferase PRDM9 guides SPO11 targeting, and the ATM kinase controls meiotic DSB numbers. Following MRE11 nuclease removal of SPO11, the DSB is resected and loaded with DMC1 filaments for homolog invasion. Here, we demonstrate the direct detection of meiotic DSBs and resection using END-seq on mouse spermatocytes with low sample input. We find that DMC1 limits both minimum and maximum resection lengths, whereas 53BP1, BRCA1 and EXO1 play surprisingly minimal roles. Through enzymatic modifications to END-seq, we identify a SPO11-bound meiotic recombination intermediate (SPO11-RI) present at all hotspots. We propose that SPO11-RI forms because chromatin-bound PRDM9 asymmetrically blocks MRE11 from releasing SPO11. In Atm-/- spermatocytes, trapped SPO11 cleavage complexes accumulate due to defective MRE11 initiation of resection. Thus, in addition to governing SPO11 breakage, ATM and PRDM9 are critical local regulators of mammalian SPO11 processing.


Asunto(s)
Endodesoxirribonucleasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Recombinación Homóloga/fisiología , Meiosis/fisiología , Espermatocitos/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Endodesoxirribonucleasas/genética , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Femenino , N-Metiltransferasa de Histona-Lisina/genética , Proteína Homóloga de MRE11/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/genética
18.
Cell Rep ; 29(13): 4471-4481.e6, 2019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31875554

RESUMEN

During V(D)J recombination, RAG proteins introduce DNA double-strand breaks (DSBs) at recombination signal sequences (RSSs) that contain either 12- or 23-nt spacer regions. Coordinated 12/23 cleavage predicts that DSBs at variable (V) gene segments should equal the level of breakage at joining (J) segments. Contrary to this, here we report abundant RAG-dependent DSBs at multiple Vκ gene segments independent of V-J rearrangement. We find that a large fraction of Vκ gene segments are flanked not only by a bone-fide 12 spacer but also an overlapping, 23-spacer flipped RSS. These compatible pairs of RSSs mediate recombination and deletion inside the Vκ cluster even in the complete absence of Jκ gene segments and support a V(D)J recombination center (RC) independent of the conventional Jκ-centered RC. We propose an improved model of Vκ-Jκ repertoire formation by incorporating these surprisingly frequent, evolutionarily conserved intra-Vκ cluster recombination events.


Asunto(s)
Linfocitos B/metabolismo , ADN/genética , Región Variable de Inmunoglobulina/genética , Recombinación V(D)J/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Sistemas CRISPR-Cas , Células Clonales , ADN/inmunología , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP)/deficiencia , ADN Ligasa (ATP)/genética , ADN Ligasa (ATP)/inmunología , Endonucleasas/deficiencia , Endonucleasas/genética , Endonucleasas/inmunología , Femenino , Edición Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Familia de Multigenes , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Bazo/citología , Bazo/inmunología
19.
Cell Rep ; 29(12): 3902-3915.e8, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31851922

RESUMEN

Somatic hypermutation (SHM) introduces point mutations into immunoglobulin (Ig) genes but also causes mutations in other parts of the genome. We have used lentiviral SHM reporter vectors to identify regions of the genome that are susceptible ("hot") and resistant ("cold") to SHM, revealing that SHM susceptibility and resistance are often properties of entire topologically associated domains (TADs). Comparison of hot and cold TADs reveals that while levels of transcription are equivalent, hot TADs are enriched for the cohesin loader NIPBL, super-enhancers, markers of paused/stalled RNA polymerase 2, and multiple important B cell transcription factors. We demonstrate that at least some hot TADs contain enhancers that possess SHM targeting activity and that insertion of a strong Ig SHM-targeting element into a cold TAD renders it hot. Our findings lead to a model for SHM susceptibility involving the cooperative action of cis-acting SHM targeting elements and the dynamic and architectural properties of TADs.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Hipermutación Somática de Inmunoglobulina/genética , Línea Celular Tumoral , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Células HEK293 , Humanos , Lentivirus , Masculino , Mutación/genética , Plásmidos/genética , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo
20.
PLoS One ; 11(2): e0149146, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26900682

RESUMEN

The activation induced cytosine deaminase (AID) mediates diversification of B cell immunoglobulin genes by the three distinct yet related processes of somatic hypermutation (SHM), class switch recombination (CSR), and gene conversion (GCV). SHM occurs in germinal center B cells, and the transcription factor Bcl6 is a key regulator of the germinal center B cell gene expression program, including expression of AID. To test the hypothesis that Bcl6 function is important for the process of SHM, we compared WT chicken DT40 B cells, which constitutively perform SHM/GCV, to their Bcl6-deficient counterparts. We found that Bcl6-deficient DT40 cells were unable to perform SHM and GCV despite enforced high level expression of AID and substantial levels of AID in the nucleus of the cells. To gain mechanistic insight into the GCV/SHM dependency on Bcl6, transcriptional features of a highly expressed SHM target gene were analyzed in Bcl6-sufficient and -deficient DT40 cells. No defect was observed in the accumulation of single stranded DNA in the target gene as a result of Bcl6 deficiency. In contrast, association of Spt5, an RNA polymerase II (Pol II) and AID binding factor, was strongly reduced at the target gene body relative to the transcription start site in Bcl6-deficient cells as compared to WT cells. However, partial reconstitution of Bcl6 function substantially reconstituted Spt5 association with the target gene body but did not restore detectable SHM. Our observations suggest that in the absence of Bcl6, Spt5 fails to associate efficiently with Pol II at SHM targets, perhaps precluding robust AID action on the SHM target DNA. Our data also suggest, however, that Spt5 binding is not sufficient for SHM of a target gene even in DT40 cells with strong expression of AID.


Asunto(s)
Pollos/genética , Proteínas de Unión al ADN/metabolismo , Conversión Génica , Hipermutación Somática de Inmunoglobulina , Factores de Transcripción/metabolismo , Animales , Línea Celular , Citidina Desaminasa/metabolismo , Proteínas de Unión al ADN/deficiencia , Expresión Génica , Técnicas de Inactivación de Genes , Genes Reporteros , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Transporte de Proteínas , Factores de Transcripción/deficiencia , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA