Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 82(6): 1107-1122.e7, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35303483

RESUMEN

Splicing factor mutations are common among cancers, recently emerging as drivers of myeloid malignancies. U2AF1 carries hotspot mutations in its RNA-binding motifs; however, how they affect splicing and promote cancer remain unclear. The U2AF1/U2AF2 heterodimer is critical for 3' splice site (3'SS) definition. To specifically unmask changes in U2AF1 function in vivo, we developed a crosslinking and immunoprecipitation procedure that detects contacts between U2AF1 and the 3'SS AG at single-nucleotide resolution. Our data reveal that the U2AF1 S34F and Q157R mutants establish new 3'SS contacts at -3 and +1 nucleotides, respectively. These effects compromise U2AF2-RNA interactions, resulting predominantly in intron retention and exon exclusion. Integrating RNA binding, splicing, and turnover data, we predicted that U2AF1 mutations directly affect stress granule components, which was corroborated by single-cell RNA-seq. Remarkably, U2AF1-mutant cell lines and patient-derived MDS/AML blasts displayed a heightened stress granule response, pointing to a novel role for biomolecular condensates in adaptive oncogenic strategies.


Asunto(s)
Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Factor de Empalme U2AF , Gránulos de Estrés , Humanos , Leucemia Mieloide Aguda/genética , Mutación , Síndromes Mielodisplásicos/genética , Sitios de Empalme de ARN , Empalme del ARN/genética , Proteínas de Unión al ARN/genética , Factor de Empalme U2AF/genética , Factor de Empalme U2AF/metabolismo , Gránulos de Estrés/metabolismo
2.
Immunity ; 52(6): 1007-1021.e8, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32497523

RESUMEN

N6-methyladenosine (m6A) is the most abundant RNA modification, but little is known about its role in mammalian hematopoietic development. Here, we show that conditional deletion of the m6A writer METTL3 in murine fetal liver resulted in hematopoietic failure and perinatal lethality. Loss of METTL3 and m6A activated an aberrant innate immune response, mediated by the formation of endogenous double-stranded RNAs (dsRNAs). The aberrantly formed dsRNAs were long, highly m6A modified in their native state, characterized by low folding energies, and predominantly protein coding. We identified coinciding activation of pattern recognition receptor pathways normally tasked with the detection of foreign dsRNAs. Disruption of the aberrant immune response via abrogation of downstream Mavs or Rnasel signaling partially rescued the observed hematopoietic defects in METTL3-deficient cells in vitro and in vivo. Our results suggest that m6A modification protects against endogenous dsRNA formation and a deleterious innate immune response during mammalian hematopoietic development.


Asunto(s)
Adenosina/química , Hematopoyesis/genética , Hematopoyesis/inmunología , Inmunidad Innata/genética , ARN Bicatenario/metabolismo , Animales , Biomarcadores , Trastornos de Fallo de la Médula Ósea/etiología , Trastornos de Fallo de la Médula Ósea/metabolismo , Trastornos de Fallo de la Médula Ósea/patología , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Epigénesis Genética , Expresión Génica , Células Madre Hematopoyéticas , Inmunofenotipificación , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Noqueados , ARN Bicatenario/química
3.
EMBO J ; 41(1): e105026, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34791698

RESUMEN

Intronic GGGGCC (G4C2) hexanucleotide repeat expansion within the human C9orf72 gene represents the most common cause of familial forms of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) (C9ALS/FTD). Repeat-associated non-AUG (RAN) translation of repeat-containing C9orf72 RNA results in the production of neurotoxic dipeptide-repeat proteins (DPRs). Here, we developed a high-throughput drug screen for the identification of positive and negative modulators of DPR levels. We found that HSP90 inhibitor geldanamycin and aldosterone antagonist spironolactone reduced DPR levels by promoting protein degradation via the proteasome and autophagy pathways respectively. Surprisingly, cAMP-elevating compounds boosting protein kinase A (PKA) activity increased DPR levels. Inhibition of PKA activity, by both pharmacological and genetic approaches, reduced DPR levels in cells and rescued pathological phenotypes in a Drosophila model of C9ALS/FTD. Moreover, knockdown of PKA-catalytic subunits correlated with reduced translation efficiency of DPRs, while the PKA inhibitor H89 reduced endogenous DPR levels in C9ALS/FTD patient-derived iPSC motor neurons. Together, our results suggest new and druggable pathways modulating DPR levels in C9ALS/FTD.


Asunto(s)
Proteína C9orf72/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Dipéptidos/metabolismo , Proteolisis , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular , Codón Iniciador/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Expansión de las Repeticiones de ADN/genética , Modelos Animales de Enfermedad , Drosophila/efectos de los fármacos , Demencia Frontotemporal/patología , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Madre Pluripotentes Inducidas/patología , Isoquinolinas/farmacología , Longevidad/efectos de los fármacos , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , Biosíntesis de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Interferencia de ARN , Sulfonamidas/farmacología
4.
Mol Cell ; 71(2): 256-270.e10, 2018 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-30029004

RESUMEN

The RNA-binding protein HuD promotes neurogenesis and favors recovery from peripheral axon injury. HuD interacts with many mRNAs, altering both stability and translation efficiency. We generated a nucleotide resolution map of the HuD RNA interactome in motor neuron-like cells, identifying HuD target sites in 1,304 mRNAs, almost exclusively in the 3' UTR. HuD binds many mRNAs encoding mTORC1-responsive ribosomal proteins and translation factors. Altered HuD expression correlates with the translation efficiency of these mRNAs and overall protein synthesis, in a mTORC1-independent fashion. The predominant HuD target is the abundant, small non-coding RNA Y3, amounting to 70% of the HuD interaction signal. Y3 functions as a molecular sponge for HuD, dynamically limiting its recruitment to polysomes and its activity as a translation and neuron differentiation enhancer. These findings uncover an alternative route to the mTORC1 pathway for translational control in motor neurons that is tunable by a small non-coding RNA.


Asunto(s)
Proteína 4 Similar a ELAV/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Neuronas Motoras/fisiología , ARN Pequeño no Traducido/genética , Regiones no Traducidas 3' , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2 , Animales , Línea Celular , Proteína 4 Similar a ELAV/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Neuronas Motoras/metabolismo , Neurogénesis/genética , Polirribosomas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Pequeño no Traducido/metabolismo
5.
J Biol Chem ; 300(2): 105648, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38219816

RESUMEN

Cellular plasticity is crucial for adapting to ever-changing stimuli. As a result, cells consistently reshape their translatome, and, consequently, their proteome. The control of translational activity has been thoroughly examined at the stage of translation initiation. However, the regulation of ribosome speed in cells is widely unknown. In this study, we utilized a timed ribosome runoff approach, along with proteomics and transmission electron microscopy, to investigate global translation kinetics in cells. We found that ribosome speeds vary among various cell types, such as astrocytes, induced pluripotent human stem cells, human neural stem cells, and human and rat neurons. Of all cell types studied, mature cortical neurons exhibit the highest rate of translation. This finding is particularly remarkable because mature cortical neurons express the eukaryotic elongation factor 2 (eEF2) at lower levels than other cell types. Neurons solve this conundrum by inactivating a fraction of their ribosomes. As a result, the increase in eEF2 levels leads to a reduction of inactive ribosomes and an enhancement of active ones. Processes that alter the demand for active ribosomes, like neuronal excitation, cause increased inactivation of redundant ribosomes in an eEF2-dependent manner. Our data suggest a novel regulatory mechanism in which neurons dynamically inactivate ribosomes to facilitate translational remodeling. These findings have important implications for developmental brain disorders characterized by, among other things, aberrant translation.


Asunto(s)
Biosíntesis de Proteínas , Ribosomas , Animales , Humanos , Ratas , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional , Ribosomas/metabolismo , Ratones , Ratones Endogámicos C57BL
6.
Nucleic Acids Res ; 51(7): 3452-3464, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-36912077

RESUMEN

Competition for intracellular resources, also known as gene expression burden, induces coupling between independently co-expressed genes, a detrimental effect on predictability and reliability of gene circuits in mammalian cells. We recently showed that microRNA (miRNA)-mediated target downregulation correlates with the upregulation of a co-expressed gene, and by exploiting miRNAs-based incoherent-feed-forward loops (iFFLs) we stabilise a gene of interest against burden. Considering these findings, we speculate that miRNA-mediated gene downregulation causes cellular resource redistribution. Despite the extensive use of miRNA in synthetic circuits regulation, this indirect effect was never reported before. Here we developed a synthetic genetic system that embeds miRNA regulation, and a mathematical model, MIRELLA, to unravel the miRNA (MI) RolE on intracellular resource aLLocAtion. We report that the link between miRNA-gene downregulation and independent genes upregulation is a result of the concerted action of ribosome redistribution and 'queueing-effect' on the RNA degradation pathway. Taken together, our results provide for the first time insights into the hidden regulatory interaction of miRNA-based synthetic networks, potentially relevant also in endogenous gene regulation. Our observations allow to define rules for complexity- and context-aware design of genetic circuits, in which transgenes co-expression can be modulated by tuning resource availability via number and location of miRNA target sites.


Asunto(s)
MicroARNs , Modelos Genéticos , Animales , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Genes Sintéticos , Mamíferos/genética , MicroARNs/genética , MicroARNs/metabolismo , Reproducibilidad de los Resultados
7.
J Cell Sci ; 135(23)2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36314272

RESUMEN

NOC1 is a nucleolar protein necessary in yeast for both transport and maturation of ribosomal subunits. Here, we show that Drosophila NOC1 (annotated CG7839) is necessary for rRNAs maturation and for a correct animal development. Its ubiquitous downregulation results in a dramatic decrease in polysome level and of protein synthesis. NOC1 expression in multiple organs, such as the prothoracic gland and the fat body, is necessary for their proper functioning. Reduction of NOC1 in epithelial cells from the imaginal discs results in clones that die by apoptosis, an event that is partially rescued in a Minute/+ background, suggesting that reduction of NOC1 induces the cells to become less fit and to acquire a 'loser' state. NOC1 downregulation activates the pro-apoptotic Eiger-JNK pathway and leads to an increase of Xrp1, which results in the upregulation of DILP8, a member of the insulin/relaxin-like family known to coordinate organ growth with animal development. Our data underline NOC1 as an essential gene in ribosome biogenesis and highlight its novel functions in the control of growth and cell competition.


Asunto(s)
Competencia Celular , Precursores del ARN , Sistema de Señalización de MAP Quinasas
8.
Biochem Soc Trans ; 52(1): 465-479, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38391004

RESUMEN

The underlying cause of Spinal Muscular Atrophy (SMA) is in the reduction of survival motor neuron (SMN) protein levels due to mutations in the SMN1 gene. The specific effects of SMN protein loss and the resulting pathological alterations are not fully understood. Given the crucial roles of the SMN protein in snRNP biogenesis and its interactions with ribosomes and translation-related proteins and mRNAs, a decrease in SMN levels below a specific threshold in SMA is expected to affect translational control of gene expression. This review covers both direct and indirect SMN interactions across various translation-related cellular compartments and processes, spanning from ribosome biogenesis to local translation and beyond. Additionally, it aims to outline deficiencies and alterations in translation observed in SMA models and patients, while also discussing the implications of the relationship between SMN protein and the translation machinery within the context of current and future therapies.


Asunto(s)
Atrofia Muscular Espinal , Humanos , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Atrofia Muscular Espinal/metabolismo , Ribosomas/metabolismo , ARN Mensajero/metabolismo , Mutación
9.
Nucleic Acids Res ; 50(4): e23, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-34850942

RESUMEN

Most RNA footprinting approaches that require ribonuclease cleavage generate RNA fragments bearing a phosphate or cyclic phosphate group at their 3' end. Unfortunately, current library preparation protocols rely only on a 3' hydroxyl group for adaptor ligation or poly-A tailing. Here, we developed circAID-p-seq, a PCR-free library preparation for selective 3' phospho-RNA sequencing. As a proof of concept, we applied circAID-p-seq to ribosome profiling, which is based on sequencing of RNA fragments protected by ribosomes after endonuclease digestion. CircAID-p-seq, combined with the dedicated computational pipeline circAidMe, facilitates accurate, fast and highly efficient sequencing of phospho-RNA fragments from eukaryotic cells and tissues. We used circAID-p-seq to portray ribosome occupancy in transcripts, providing a versatile and PCR-free strategy to possibly unravel any endogenous 3'-phospho RNA molecules.


Asunto(s)
ARN , Ribosomas , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Fosfatos , ARN/genética , Ribosomas/genética , Análisis de Secuencia de ARN/métodos
10.
RNA Biol ; 18(6): 914-931, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33043783

RESUMEN

Previous high-throughput studies in Gram-negative bacteria identified a large number of 3'UTR fragments that potentially function as sRNAs. Here we extensively characterize the MalH sRNA. We show that MalH is a stable degradation intermediate derived from the 3' end of malG, which is part of the maltose uptake operon transcript malEFG. Unlike the majority of bacterial sRNAs, MalH is transiently expressed during the transition from the exponential to the stationary growth phase, suggesting that it contributes to adaptation to changes in nutrient availability. Over-expression of MalH reduces expression of general outer membrane porins and MicA, a repressor of the high-affinity maltose/maltodextrin transporter LamB. Disrupting MalH production and function significantly reduces lamB accumulation when maltose is the only available carbon source, presumably due to the accumulation of the MicA repressor. We propose that MalH is part of a regulatory network that, during the transition phase, directly or indirectly promotes accumulation of high-affinity maltose transporters in the outer membrane by dampening competing pathways.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Carbono/metabolismo , Proteínas de Escherichia coli/genética , Porinas/genética , ARN Bacteriano/genética , ARN Mensajero/genética , ARN Pequeño no Traducido/genética , Receptores Virales/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Bacteriana de la Expresión Génica , Maltosa/metabolismo , Operón/genética , Porinas/metabolismo , Unión Proteica , ARN Bacteriano/metabolismo , ARN Mensajero/metabolismo , ARN Pequeño no Traducido/metabolismo , RNA-Seq/métodos , Receptores Virales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
11.
Hum Mol Genet ; 27(16): 2851-2862, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29790918

RESUMEN

Spinal muscular atrophy (SMA) is a progressive motor neuron disease caused by deleterious variants in SMN1 that lead to a marked decrease in survival motor neuron (SMN) protein expression. Humans have a second SMN gene (SMN2) that is almost identical to SMN1. However, due to alternative splicing the majority of SMN2 messenger ribonucleic acid (mRNA) is translated into a truncated, unstable protein that is quickly degraded. Because the presence of SMN2 provides a unique opportunity for therapy development in SMA patients, the mechanisms that regulate SMN2 splicing and mRNA expression have been elucidated in great detail. In contrast, how much SMN protein is produced at different developmental time points and in different tissues remains under-characterized. In this study, we addressed this issue by determining SMN protein expression levels at three developmental time points across six different mouse tissues and in two distinct mouse models of SMA ('severe' Taiwanese and 'intermediate' Smn2B/- mice). We found that, in healthy control mice, SMN protein expression was significantly influenced by both age and tissue type. When comparing mouse models of SMA, we found that, despite being transcribed from genetically different alleles, control SMN levels were relatively similar. In contrast, the degree of SMN depletion between tissues in SMA varied substantially over time and between the two models. These findings offer an explanation for the differential vulnerability of tissues and organs observed in SMA and further our understanding of the systemic and temporal requirements for SMN with direct relevance for developing effective therapies for SMA.


Asunto(s)
Atrofia Muscular Espinal/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Empalme Alternativo/genética , Animales , Modelos Animales de Enfermedad , Exones , Humanos , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/fisiopatología , Empalme del ARN/genética , Médula Espinal/fisiopatología , Proteína 2 para la Supervivencia de la Neurona Motora/genética
12.
RNA ; 24(7): 915-925, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29643068

RESUMEN

Schwann cells are key players in neuro-regeneration: They sense "alarm" signals released by degenerating nerve terminals and differentiate toward a proregenerative phenotype, with phagocytosis of nerve debris and nerve guidance. At the murine neuromuscular junction, hydrogen peroxide (H2O2) is a key signal of Schwann cells' activation in response to a variety of nerve injuries. Here we report that Schwann cells exposed to low doses of H2O2 rewire the expression of several RNAs at both transcriptional and translational levels. Among the genes positively regulated at both levels, we identified an enriched cluster involved in cytoskeleton remodeling and cell migration, with the Annexin (Anxa) proteins being the most represented family. We show that both Annexin A2 (Anxa2) transcript and protein accumulate at the tips of long pseudopods that Schwann cells extend upon H2O2 exposure. Interestingly, Schwann cells reply to this signal and to nerve injury by locally translating Anxa2 in pseudopods, and undergo an extensive cytoskeleton remodeling. Our results show that, similarly to neurons, Schwann cells take advantage of local protein synthesis to change shape and move toward damaged axonal terminals to facilitate axonal regeneration.


Asunto(s)
Anexina A2/biosíntesis , Peróxido de Hidrógeno/farmacología , Células de Schwann/metabolismo , Animales , Anexina A2/genética , Anexina A2/metabolismo , Células Cultivadas , Citoesqueleto/ultraestructura , Regulación de la Expresión Génica/efectos de los fármacos , Ratones Endogámicos C57BL , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Biosíntesis de Proteínas , ARN/biosíntesis , Células de Schwann/citología , Células de Schwann/efectos de los fármacos , Células de Schwann/ultraestructura , Transcriptoma/efectos de los fármacos
13.
PLoS Comput Biol ; 14(8): e1006169, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30102689

RESUMEN

Ribosome profiling is a powerful technique used to study translation at the genome-wide level, generating unique information concerning ribosome positions along RNAs. Optimal localization of ribosomes requires the proper identification of the ribosome P-site in each ribosome protected fragment, a crucial step to determine the trinucleotide periodicity of translating ribosomes, and draw correct conclusions concerning where ribosomes are located. To determine the P-site within ribosome footprints at nucleotide resolution, the precise estimation of its offset with respect to the protected fragment is necessary. Here we present riboWaltz, an R package for calculation of optimal P-site offsets, diagnostic analysis and visual inspection of ribosome profiling data. Compared to existing tools, riboWaltz shows improved accuracies for P-site estimation and neat ribosome positioning in multiple case studies. riboWaltz was implemented in R and is available as an R package at https://github.com/LabTranslationalArchitectomics/RiboWaltz.


Asunto(s)
Biología Computacional/métodos , Ribosomas/fisiología , Análisis de Secuencia de ARN/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Biosíntesis de Proteínas/fisiología , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Programas Informáticos
14.
Nucleic Acids Res ; 45(11): 6775-6792, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28379492

RESUMEN

RALY is a member of the heterogeneous nuclear ribonucleoprotein family (hnRNP), a large family of RNA-binding proteins involved in many aspects of RNA metabolism. Although RALY interactome has been recently characterized, a comprehensive global analysis of RALY-associated RNAs is lacking and the biological function of RALY remains elusive. Here, we performed RIP-seq analysis to identify RALY interacting RNAs and assessed the role of RALY in gene expression. We demonstrate that RALY binds specific coding and non-coding RNAs and associates with translating mRNAs of mammalian cells. Among the identified transcripts, we focused on ANXA1 and H1FX mRNAs, encoding for Annexin A1 and for the linker variant of the histone H1X, respectively. Both proteins are differentially expressed by proliferating cells and are considered as markers for tumorigenesis. We demonstrate that cells lacking RALY expression exhibit changes in the levels of H1FX and ANXA1 mRNAs and proteins in an opposite manner. We also provide evidence for a direct binding of RALY to the U-rich elements present within the 3΄UTR of both transcripts. Thus, our results identify RALY as a poly-U binding protein and as a regulator of H1FX and ANXA1 in mammalian cells.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo C/fisiología , ARN Mensajero/metabolismo , Regiones no Traducidas 3' , Anexina A1/genética , Anexina A1/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Ciclo Celular , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Células MCF-7 , Polirribosomas/metabolismo , Unión Proteica
15.
PLoS Pathog ; 12(4): e1005528, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27055274

RESUMEN

Bacterial genotoxins, produced by several Gram-negative bacteria, induce DNA damage in the target cells. While the responses induced in the host cells have been extensively studied in vitro, the role of these effectors during the course of infection remains poorly characterized. To address this issue, we assessed the effects of the Salmonella enterica genotoxin, known as typhoid toxin, in in vivo models of murine infection. Immunocompetent mice were infected with isogenic S. enterica, serovar Typhimurium (S. Typhimurium) strains, encoding either a functional or an inactive typhoid toxin. The presence of the genotoxic subunit was detected 10 days post-infection in the liver of infected mice. Unexpectedly, its expression promoted the survival of the host, and was associated with a significant reduction of severe enteritis in the early phases of infection. Immunohistochemical and transcriptomic analysis confirmed the toxin-mediated suppression of the intestinal inflammatory response. The presence of a functional typhoid toxin further induced an increased frequency of asymptomatic carriers. Our data indicate that the typhoid toxin DNA damaging activity increases host survival and favours long-term colonization, highlighting a complex cross-talk between infection, DNA damage response and host immune response. These findings may contribute to understand why such effectors have been evolutionary conserved and horizontally transferred among Gram-negative bacteria.


Asunto(s)
Infecciones Asintomáticas , Enfermedades Transmisibles/microbiología , Mutágenos/toxicidad , Salmonella typhimurium/patogenicidad , Fiebre Tifoidea/microbiología , Animales , Intestinos/microbiología , Macrófagos/microbiología , Ratones , Virulencia
16.
Nucleic Acids Res ; 43(22): e153, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26240374

RESUMEN

Fluctuations in mRNA levels only partially contribute to determine variations in mRNA availability for translation, producing the well-known poor correlation between transcriptome and proteome data. Recent advances in microscopy now enable researchers to obtain high resolution images of ribosomes on transcripts, providing precious snapshots of translation in vivo. Here we propose RiboAbacus, a mathematical model that for the first time incorporates imaging data in a predictive model of transcript-specific ribosome densities and translational efficiencies. RiboAbacus uses a mechanistic model of ribosome dynamics, enabling the quantification of the relative importance of different features (such as codon usage and the 5' ramp effect) in determining the accuracy of predictions. The model has been optimized in the human Hek-293 cell line to fit thousands of images of human polysomes obtained by atomic force microscopy, from which we could get a reference distribution of the number of ribosomes per mRNA with unmatched resolution. After validation, we applied RiboAbacus to three case studies of known transcriptome-proteome datasets for estimating the translational efficiencies, resulting in an increased correlation with corresponding proteomes. RiboAbacus is an intuitive tool that allows an immediate estimation of crucial translation properties for entire transcriptomes, based on easily obtainable transcript expression levels.


Asunto(s)
Modelos Biológicos , Polirribosomas/ultraestructura , Biosíntesis de Proteínas , Transcriptoma , Animales , Células HEK293 , Humanos , Células MCF-7 , Microscopía de Fuerza Atómica , Proteómica , Conejos , Reticulocitos/ultraestructura , Ribosomas/ultraestructura , Programas Informáticos
17.
Biochem J ; 472(1): 83-95, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26371376

RESUMEN

Genome-wide analyses of translation can provide major contributions in our understanding of the complex interplay between virulent factors and host cells. So far, the activation of host translational control mechanisms by bacterial toxins, owing to specific recruitment of mRNAs, RNA-binding proteins (RBPs) and ncRNAs (non-coding RNAs), are far from being understood. In the present study, we characterize for the first time the changes experienced by the translational control system of host cells in response to the well-known Staphylococcus aureus α-haemolysin (AHL) under both sublytic and lytic conditions. By comparing variations occurring in the cellular transcriptome and translatome, we give evidence that global gene expression is primarily rewired at the translational level, with the contribution of the RBP ELAVL1 (HuR) in the sublytic response. These results reveal the importance of translational control during host-pathogen interaction, opening new approaches for AHL-induced diseases.


Asunto(s)
Toxinas Bacterianas/farmacología , Variación Genética/efectos de los fármacos , Proteínas Hemolisinas/farmacología , Biosíntesis de Proteínas/genética , Transcriptoma/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Proteína 1 Similar a ELAV/genética , Proteína 1 Similar a ELAV/metabolismo , Perfilación de la Expresión Génica/métodos , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Immunoblotting , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Bioinformatics ; 30(2): 289-91, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24222209

RESUMEN

UNLABELLED: High-throughput technologies have led to an explosion of genomic data available for automated analysis. The consequent possibility to simultaneously sample multiple layers of variation along the gene expression flow requires computational methods integrating raw information from different '-omics'. It has been recently demonstrated that translational control is a widespread phenomenon, with profound and still underestimated regulation capabilities. Although detecting changes in the levels of total messenger RNAs (mRNAs; the transcriptome), of polysomally loaded mRNAs (the translatome) and of proteins (the proteome) is experimentally feasible in a high-throughput way, the integration of these levels is still far from being robustly approached. Here we introduce tRanslatome, a new R/Bioconductor package, which is a complete platform for the simultaneous pairwise analysis of transcriptome, translatome and proteome data. The package includes most of the available statistical methods developed for the analysis of high-throughput data, allowing the parallel comparison of differentially expressed genes and the corresponding differentially enriched biological themes. Notably, it also enables the prediction of translational regulatory elements on mRNA sequences. The utility of this tool is demonstrated with two case studies. AVAILABILITY AND IMPLEMENTATION: tRanslatome is available in Bioconductor.


Asunto(s)
Biología Computacional , Biosíntesis de Proteínas , Proteoma/análisis , Secuencias Reguladoras de Ácidos Nucleicos/genética , Programas Informáticos , Transcriptoma , Diferenciación Celular , Bases de Datos Factuales , Genómica , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , ARN Mensajero/genética
19.
Subcell Biochem ; 80: 271-91, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24798017

RESUMEN

Proteins with membrane-attack complex/perforin (MACPF) domains are found in almost all kingdoms of life, and they have a variety of biological roles, including defence and attack, organism development, and cell adhesion and signalling. The distribution of these proteins in fungi appears to be restricted to some Pezizomycotina and Basidiomycota species only, in correlation with another group of proteins with unknown biological function, known as aegerolysins. These two protein groups coincide in only a few species, and they might operate in concert as cytolytic bi-component pore-forming agents. Representative proteins here include pleurotolysin B, which has a MACPF domain, and the aegerolysin-like protein pleurotolysin A, and the very similar ostreolysin A, which have been purified from oyster mushroom (Pleurotus ostreatus). These have been shown to act in concert to perforate natural and artificial lipid membranes with high cholesterol and sphingomyelin content. The aegerolysin-like proteins provide the membrane cholesterol/sphingomyelin selectivity and recruit oligomerised pleurotolysin B molecules, to create a membrane-inserted pore complex. The resulting protein structure has been imaged with electron microscopy, and it has a 13-meric rosette-like structure, with a central lumen that is ~4-5 nm in diameter. The opened transmembrane pore is non-selectively permeable for ions and smaller neutral solutes, and is a cause of cytolysis of a colloid-osmotic type. The biological significance of these proteins for the fungal life-style is discussed.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Proteínas Fúngicas/fisiología , Proteínas Hemolisinas/fisiología , Perforina/fisiología , Proteínas Citotóxicas Formadoras de Poros/fisiología , Secuencia de Aminoácidos , Animales , Complejo de Ataque a Membrana del Sistema Complemento/química , Proteínas Fúngicas/química , Proteínas Hemolisinas/química , Humanos , Datos de Secuencia Molecular , Perforina/química , Filogenia , Pleurotus/genética , Pleurotus/patogenicidad , Proteínas Citotóxicas Formadoras de Poros/química , Multimerización de Proteína/fisiología , Homología de Secuencia de Aminoácido
20.
J Biol Chem ; 288(33): 23704-15, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23803608

RESUMEN

Actinoporin equinatoxin II (EqtII) is an archetypal example of α-helical pore-forming toxins that porate cellular membranes by the use of α-helices. Previous studies proposed several steps in the pore formation: binding of monomeric protein onto the membrane, followed by oligomerization and insertion of the N-terminal α-helix into the lipid bilayer. We studied these separate steps with an EqtII triple cysteine mutant. The mutant was engineered to monitor the insertion of the N terminus into the lipid bilayer by labeling Cys-18 with a fluorescence probe and at the same time to control the flexibility of the N-terminal region by the disulfide bond formed between cysteines introduced at positions 8 and 69. The insertion of the N terminus into the membrane proceeded shortly after the toxin binding and was followed by oligomerization. The oxidized, non-lytic, form of the mutant was still able to bind to membranes and oligomerize at the same level as the wild-type or the reduced form. However, the kinetics of the N-terminal helix insertion, the release of calcein from erythrocyte ghosts, and hemolysis of erythrocytes was much slower when membrane-bound oxidized mutant was reduced by the addition of the reductant. Results show that the N-terminal region needs to be inserted in the lipid membrane before the oligomerization into the final pore and imply that there is no need for a stable prepore formation. This is different from ß-pore-forming toxins that often form ß-barrel pores via a stable prepore complex.


Asunto(s)
Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Venenos de Cnidarios/química , Venenos de Cnidarios/farmacología , Animales , Bovinos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cisteína/genética , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Cinética , Lípidos de la Membrana/química , Modelos Biológicos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ingeniería de Proteínas , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Espectrometría de Fluorescencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA