Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 207(8): 998-1011, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36724365

RESUMEN

Rationale: Chronic obstructive pulmonary disease (COPD) is a disease characterized by persistent airway inflammation and disordered macrophage function. The extent to which alterations in macrophage bioenergetics contribute to impaired antioxidant responses and disease pathogenesis has yet to be fully delineated. Objectives: Through the study of COPD alveolar macrophages (AMs) and peripheral monocyte-derived macrophages (MDMs), we sought to establish if intrinsic defects in core metabolic processes drive macrophage dysfunction and redox imbalance. Methods: AMs and MDMs from donors with COPD and healthy donors underwent functional, metabolic, and transcriptional profiling. Measurements and Main Results: We observed that AMs and MDMs from donors with COPD display a critical depletion in glycolytic- and mitochondrial respiration-derived energy reserves and an overreliance on glycolysis as a source for ATP, resulting in reduced energy status. Defects in oxidative metabolism extend to an impaired redox balance associated with defective expression of the NADPH-generating enzyme, ME1 (malic enzyme 1), a known target of the antioxidant transcription factor NRF2 (nuclear factor erythroid 2-related factor 2). Consequently, selective activation of NRF2 resets the COPD transcriptome, resulting in increased generation of TCA cycle intermediaries, improved energetic status, favorable redox balance, and recovery of macrophage function. Conclusions: In COPD, an inherent loss of metabolic plasticity leads to metabolic exhaustion and reduced redox capacity, which can be rescued by activation of the NRF2 pathway. Targeting these defects, via NRF2 augmentation, may therefore present an attractive therapeutic strategy for the treatment of the aberrant airway inflammation described in COPD.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Macrófagos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Malato Deshidrogenasa/metabolismo
2.
Bioinformatics ; 38(21): 4927-4933, 2022 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-36094347

RESUMEN

MOTIVATION: A common experimental output in biomedical science is a list of genes implicated in a given biological process or disease. The gene lists resulting from a group of studies answering the same, or similar, questions can be combined by ranking aggregation methods to find a consensus or a more reliable answer. Evaluating a ranking aggregation method on a specific type of data before using it is required to support the reliability since the property of a dataset can influence the performance of an algorithm. Such evaluation on gene lists is usually based on a simulated database because of the lack of a known truth for real data. However, simulated datasets tend to be too small compared to experimental data and neglect key features, including heterogeneity of quality, relevance and the inclusion of unranked lists. RESULTS: In this study, a group of existing methods and their variations that are suitable for meta-analysis of gene lists are compared using simulated and real data. Simulated data were used to explore the performance of the aggregation methods as a function of emulating the common scenarios of real genomic data, with various heterogeneity of quality, noise level and a mix of unranked and ranked data using 20 000 possible entities. In addition to the evaluation with simulated data, a comparison using real genomic data on the SARS-CoV-2 virus, cancer (non-small cell lung cancer) and bacteria (macrophage apoptosis) was performed. We summarize the results of our evaluation in a simple flowchart to select a ranking aggregation method, and in an automated implementation using the meta-analysis by information content algorithm to infer heterogeneity of data quality across input datasets. AVAILABILITY AND IMPLEMENTATION: The code for simulated data generation and running edited version of algorithms: https://github.com/baillielab/comparison_of_RA_methods. Code to perform an optimal selection of methods based on the results of this review, using the MAIC algorithm to infer the characteristics of an input dataset, can be downloaded here: https://github.com/baillielab/maic. An online service for running MAIC: https://baillielab.net/maic. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
COVID-19 , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Algoritmos , Carcinoma de Pulmón de Células no Pequeñas/genética , COVID-19/genética , Neoplasias Pulmonares/genética , Reproducibilidad de los Resultados , SARS-CoV-2 , Metaanálisis como Asunto
4.
Nat Immunol ; 23(6): 927-939, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35624205

RESUMEN

Hypoxemia is a defining feature of acute respiratory distress syndrome (ARDS), an often-fatal complication of pulmonary or systemic inflammation, yet the resulting tissue hypoxia, and its impact on immune responses, is often neglected. In the present study, we have shown that ARDS patients were hypoxemic and monocytopenic within the first 48 h of ventilation. Monocytopenia was also observed in mouse models of hypoxic acute lung injury, in which hypoxemia drove the suppression of type I interferon signaling in the bone marrow. This impaired monopoiesis resulted in reduced accumulation of monocyte-derived macrophages and enhanced neutrophil-mediated inflammation in the lung. Administration of colony-stimulating factor 1 in mice with hypoxic lung injury rescued the monocytopenia, altered the phenotype of circulating monocytes, increased monocyte-derived macrophages in the lung and limited injury. Thus, tissue hypoxia altered the dynamics of the immune response to the detriment of the host and interventions to address the aberrant response offer new therapeutic strategies for ARDS.


Asunto(s)
Lesión Pulmonar , Síndrome de Dificultad Respiratoria , Animales , Humanos , Hipoxia/etiología , Inflamación/complicaciones , Pulmón , Lesión Pulmonar/complicaciones , Ratones
5.
Curr Res Transl Med ; 70(2): 103333, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35104687

RESUMEN

BACKGROUND: The human protein transmembrane protease serine type 2 (TMPRSS2) plays a key role in SARS-CoV-2 infection, as it is required to activate the virus' spike protein, facilitating entry into target cells. We hypothesized that naturally-occurring TMPRSS2 human genetic variants affecting the structure and function of the TMPRSS2 protein may modulate the severity of SARS-CoV-2 infection. METHODS: We focused on the only common TMPRSS2 non-synonymous variant predicted to be damaging (rs12329760 C>T, p.V160M), which has a minor allele frequency ranging from 0.14 in Ashkenazi Jewish to 0.38 in East Asians. We analysed the association between the rs12329760 and COVID-19 severity in 2,244 critically ill patients with COVID-19 from 208 UK intensive care units recruited as part of the GenOMICC (Genetics Of Mortality In Critical Care) study. Logistic regression analyses were adjusted for sex, age and deprivation index. For in vitro studies, HEK293 cells were co-transfected with ACE2 and either TMPRSS2 wild type or mutant (TMPRSS2V160M). A SARS-CoV-2 pseudovirus entry assay was used to investigate the ability of TMPRSS2V160M to promote viral entry. RESULTS: We show that the T allele of rs12329760 is associated with a reduced likelihood of developing severe COVID-19 (OR 0.87, 95%CI:0.79-0.97, p = 0.01). This association was stronger in homozygous individuals when compared to the general population (OR 0.65, 95%CI:0.50-0.84, p = 1.3 × 10-3). We demonstrate in vitro that this variant, which causes the amino acid substitution valine to methionine, affects the catalytic activity of TMPRSS2 and is less able to support SARS-CoV-2 spike-mediated entry into cells. CONCLUSION: TMPRSS2 rs12329760 is a common variant associated with a significantly decreased risk of severe COVID-19. Further studies are needed to assess the expression of TMPRSS2 across different age groups. Moreover, our results identify TMPRSS2 as a promising drug target, with a potential role for camostat mesilate, a drug approved for the treatment of chronic pancreatitis and postoperative reflux esophagitis, in the treatment of COVID-19. Clinical trials are needed to confirm this.


Asunto(s)
COVID-19 , COVID-19/genética , Frecuencia de los Genes , Células HEK293 , Humanos , SARS-CoV-2 , Serina Endopeptidasas/genética , Internalización del Virus
6.
Viruses ; 13(8)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34452438

RESUMEN

Herpesviruses are large DNA viruses, which encode up to 300 different proteins including enzymes enabling efficient replication. Nevertheless, they depend on a multitude of host cell proteins for successful propagation. To uncover cellular host factors important for replication of pseudorabies virus (PrV), an alphaherpesvirus of swine, we performed an unbiased genome-wide CRISPR/Cas9 forward screen. To this end, a porcine CRISPR-knockout sgRNA library (SsCRISPRko.v1) targeting 20,598 genes was generated and used to transduce porcine kidney cells. Cells were then infected with either wildtype PrV (PrV-Ka) or a PrV mutant (PrV-gD-Pass) lacking the receptor-binding protein gD, which regained infectivity after serial passaging in cell culture. While no cells survived infection with PrV-Ka, resistant cell colonies were observed after infection with PrV-gD-Pass. In these cells, sphingomyelin synthase 1 (SMS1) was identified as the top hit candidate. Infection efficiency was reduced by up to 90% for PrV-gD-Pass in rabbit RK13-sgms1KO cells compared to wildtype cells accompanied by lower viral progeny titers. Exogenous expression of SMS1 partly reverted the entry defect of PrV-gD-Pass. In contrast, infectivity of PrV-Ka was reduced by 50% on the knockout cells, which could not be restored by exogenous expression of SMS1. These data suggest that SMS1 plays a pivotal role for PrV infection, when the gD-mediated entry pathway is blocked.


Asunto(s)
Sistemas CRISPR-Cas/genética , Genoma Viral , Herpesvirus Suido 1/genética , Interacciones Microbiota-Huesped , Mutación , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Animales , Línea Celular , Edición Génica , Riñón/citología , Riñón/virología , Porcinos , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Replicación Viral
7.
Sci Rep ; 10(1): 22303, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33339864

RESUMEN

The increasing body of literature describing the role of host factors in COVID-19 pathogenesis demonstrates the need to combine diverse, multi-omic data to evaluate and substantiate the most robust evidence and inform development of therapies. Here we present a dynamic ranking of host genes implicated in human betacoronavirus infection (SARS-CoV-2, SARS-CoV, MERS-CoV, seasonal coronaviruses). We conducted an extensive systematic review of experiments identifying potential host factors. Gene lists from diverse sources were integrated using Meta-Analysis by Information Content (MAIC). This previously described algorithm uses data-driven gene list weightings to produce a comprehensive ranked list of implicated host genes. From 32 datasets, the top ranked gene was PPIA, encoding cyclophilin A, a druggable target using cyclosporine. Other highly-ranked genes included proposed prognostic factors (CXCL10, CD4, CD3E) and investigational therapeutic targets (IL1A) for COVID-19. Gene rankings also inform the interpretation of COVID-19 GWAS results, implicating FYCO1 over other nearby genes in a disease-associated locus on chromosome 3. Researchers can search and review the gene rankings and the contribution of different experimental methods to gene rank at https://baillielab.net/maic/covid19 . As new data are published we will regularly update the list of genes as a resource to inform and prioritise future studies.


Asunto(s)
COVID-19/epidemiología , COVID-19/genética , Algoritmos , Complejo CD3/genética , Antígenos CD4/genética , Quimiocina CXCL10/genética , Biología Computacional , Ciclofilina A/genética , Ciclosporina/farmacología , Bases de Datos Genéticas , Estudio de Asociación del Genoma Completo , Genómica , Humanos , Sistema Inmunológico , Inmunogenética , Inflamación , Interleucina-1alfa/genética , Proteínas Asociadas a Microtúbulos/genética , Proteómica
8.
J Virol ; 94(10)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32161175

RESUMEN

Macrophages in the lung detect and respond to influenza A virus (IAV), determining the nature of the immune response. Using terminal-depth cap analysis of gene expression (CAGE), we quantified transcriptional activity of both host and pathogen over a 24-h time course of IAV infection in primary human monocyte-derived macrophages (MDMs). This method allowed us to observe heterogenous host sequences incorporated into IAV mRNA, "snatched" 5' RNA caps, and corresponding RNA sequences from host RNAs. In order to determine whether cap-snatching is random or exhibits a bias, we systematically compared host sequences incorporated into viral mRNA ("snatched") against a complete survey of all background host RNA in the same cells, at the same time. Using a computational strategy designed to eliminate sources of bias due to read length, sequencing depth, and multimapping, we were able to quantify overrepresentation of host RNA features among the sequences that were snatched by IAV. We demonstrate biased snatching of numerous host RNAs, particularly small nuclear RNAs (snRNAs), and avoidance of host transcripts encoding host ribosomal proteins, which are required by IAV for replication. We then used a systems approach to describe the transcriptional landscape of the host response to IAV, observing many new features, including a failure of IAV-treated MDMs to induce feedback inhibitors of inflammation, seen in response to other treatments.IMPORTANCE Infection with influenza A virus (IAV) infection is responsible for an estimated 500,000 deaths and up to 5 million cases of severe respiratory illness each year. In this study, we looked at human primary immune cells (macrophages) infected with IAV. Our method allows us to look at both the host and the virus in parallel. We used these data to explore a process known as "cap-snatching," where IAV snatches a short nucleotide sequence from capped host RNA. This process was believed to be random. We demonstrate biased snatching of numerous host RNAs, including those associated with snRNA transcription, and avoidance of host transcripts encoding host ribosomal proteins, which are required by IAV for replication. We then describe the transcriptional landscape of the host response to IAV, observing new features, including a failure of IAV-treated MDMs to induce feedback inhibitors of inflammation, seen in response to other treatments.


Asunto(s)
Secuencia de Bases , Virus de la Influenza A/genética , Gripe Humana/virología , Transcripción Genética/fisiología , Sesgo , Redes Reguladoras de Genes , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Virus de la Influenza A/efectos de los fármacos , Lipopolisacáridos/farmacología , Macrófagos , Caperuzas de ARN/genética , ARN Mensajero , ARN Nuclear Pequeño/metabolismo , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , Replicación Viral
9.
Nat Commun ; 11(1): 164, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31919360

RESUMEN

Host dependency factors that are required for influenza A virus infection may serve as therapeutic targets as the virus is less likely to bypass them under drug-mediated selection pressure. Previous attempts to identify host factors have produced largely divergent results, with few overlapping hits across different studies. Here, we perform a genome-wide CRISPR/Cas9 screen and devise a new approach, meta-analysis by information content (MAIC) to systematically combine our results with prior evidence for influenza host factors. MAIC out-performs other meta-analysis methods when using our CRISPR screen as validation data. We validate the host factors, WDR7, CCDC115 and TMEM199, demonstrating that these genes are essential for viral entry and regulation of V-type ATPase assembly. We also find that CMTR1, a human mRNA cap methyltransferase, is required for efficient viral cap snatching and regulation of a cell autonomous immune response, and provides synergistic protection with the influenza endonuclease inhibitor Xofluza.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Interacciones Huésped-Patógeno/genética , Virus de la Influenza A/patogenicidad , Gripe Humana/genética , Gripe Humana/patología , Células A549 , Proteínas Adaptadoras Transductoras de Señales/genética , Antivirales/farmacología , Sistemas CRISPR-Cas , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Dibenzotiepinas , Estudio de Asociación del Genoma Completo , Humanos , Proteínas de la Membrana/genética , Metiltransferasas/metabolismo , Morfolinas , Proteínas del Tejido Nervioso/genética , Oxazinas/farmacología , Piridinas/farmacología , Piridonas , Tiepinas/farmacología , Triazinas/farmacología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Internalización del Virus
10.
J Vet Intern Med ; 32(2): 805-814, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29460300

RESUMEN

BACKGROUND: Venous air embolism is a potentially life-threatening complication of IV catheter use in horses. Despite widespread anecdotal reports of their occurrence, few cases have been reported in the literature and the prognosis is currently unknown. HYPOTHESIS/OBJECTIVES: Our objective was to describe the surrounding circumstances, clinical signs, treatment, progression, and outcome of venous air embolism in hospitalized horses. ANIMALS: Thirty-two horses with acute onset of compatible clinical signs associated with IV catheter disconnection or damage. METHODS: Multicenter retrospective study. Data extracted from clinical records included signalment, presenting complaint, catheter details, clinical signs, treatments, and outcome. RESULTS: Most cases resulted from extension set disconnection occurring within approximately 24 hours after catheter placement. In fewer horses, extension set damage was cited as a cause. Common clinical signs included tachycardia, tachypnea, recumbency, muscle fasciculations and agitation, with abnormal behavior including kicking and flank biting. Less commonly, pathological arrhythmias or more severe neurologic signs, including blindness and seizures, were noted. Progression was unpredictable, with some affected horses developing delayed-onset neurologic signs. Mortality was 6/32 (19%), including 2 cases of sudden death and other horses euthanized because of persistent neurologic deficits. Negative outcomes were more common in horses with recorded blindness, sweating or recumbency, but blindness resolved in 5/8 affected horses. CONCLUSIONS AND CLINICAL IMPORTANCE: The prognosis for resolution of clinical signs after air embolism is fair, but permanent neurologic deficits or pathologic cardiac arrhythmias can arise. Unpredictable progression warrants close monitoring. Systematic clinic-based surveillance could provide additional useful information to aid prevention.


Asunto(s)
Embolia Aérea/veterinaria , Enfermedades de los Caballos/etiología , Dispositivos de Acceso Vascular/veterinaria , Animales , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/veterinaria , Ceguera/complicaciones , Ceguera/veterinaria , Embolia Aérea/complicaciones , Embolia Aérea/etiología , Embolia Aérea/mortalidad , Enfermedades de los Caballos/mortalidad , Caballos , Enfermedades del Sistema Nervioso/complicaciones , Enfermedades del Sistema Nervioso/veterinaria , Estudios Retrospectivos , Convulsiones/complicaciones , Convulsiones/veterinaria , Dispositivos de Acceso Vascular/efectos adversos
11.
J Allergy Clin Immunol ; 141(1): 152-162, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28583370

RESUMEN

BACKGROUND: Atopic dermatitis (AD) and allergic contact dermatitis (ACD) are both forms of eczema and are common inflammatory skin diseases with a central role of T cell-derived IL-22 in their pathogenesis. Although prostaglandin (PG) E2 is known to promote inflammation, little is known about its role in processes related to AD and ACD development, including IL-22 upregulation. OBJECTIVES: We sought to investigate whether PGE2 has a role in IL-22 induction and development of ACD, which has increased prevalence in patients with AD. METHODS: T-cell cultures and in vivo sensitization of mice with haptens were used to assess the role of PGE2 in IL-22 production. The involvement of PGE2 receptors and their downstream signals was also examined. The effects of PGE2 were evaluated by using the oxazolone-induced ACD mouse model. The relationship of PGE2 and IL-22 signaling pathways in skin inflammation were also investigated by using genomic profiling in human lesional AD skin. RESULTS: PGE2 induces IL-22 from T cells through its receptors, E prostanoid receptor (EP) 2 and EP4, and involves cyclic AMP signaling. Selective deletion of EP4 in T cells prevents hapten-induced IL-22 production in vivo, and limits atopic-like skin inflammation in the oxazolone-induced ACD model. Moreover, both PGE2 and IL-22 pathway genes were coordinately upregulated in human AD lesional skin but were at less than significant detection levels after corticosteroid or UVB treatments. CONCLUSIONS: Our results define a crucial role for PGE2 in promoting ACD by facilitating IL-22 production from T cells.


Asunto(s)
Dermatitis Alérgica por Contacto/inmunología , Dinoprostona/inmunología , Interleucinas/inmunología , Piel/inmunología , Linfocitos T/inmunología , Animales , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/patología , Dinoprostona/genética , Humanos , Interleucinas/genética , Ratones , Ratones Noqueados , Piel/patología , Linfocitos T/patología , Interleucina-22
12.
PLoS One ; 12(5): e0177664, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28552958

RESUMEN

The innate immune response to lipopolysaccharide contributes substantially to the morbidity and mortality of gram-negative sepsis. Horses and humans share an exquisite sensitivity to lipopolysaccharide and thus the horse may provide valuable comparative insights into this aspect of the inflammatory response. MicroRNAs, small non-coding RNA molecules acting as post-transcriptional regulators of gene expression, have key roles in toll-like receptor signaling regulation but have not been studied in this context in horses. The central hypothesis of this study was that lipopolysaccharide induces differential microRNA expression in equine peripheral blood mononuclear cells in a manner comparable to humans. Illumina Next Generation Sequencing was used to characterize the basal microRNA transcriptome in isolated peripheral blood mononuclear cells from healthy adult horses, and to evaluate LPS-induced changes in microRNA expression in cells cultured for up to four hours. Selected expression changes were validated using quantitative reverse-transcriptase PCR. Only miR-155 was significantly upregulated by LPS, changing in parallel with supernatant tumor necrosis factor-α concentration. Eight additional microRNAs, including miR-146a and miR-146b, showed significant expression change with time in culture without a clear LPS effect. Target predictions indicated a number of potential immunity-associated targets for miR-155 in the horse, including SOCS1, TAB2 and elements of the PI3K signaling pathway, suggesting that it is likely to influence the acute inflammatory response to LPS. Gene alignment showed extensive conservation of the miR-155 precursor gene and associated promoter regions between horses and humans. The basal and LPS-stimulated microRNA expression pattern characterized here were similar to those described in human leukocytes. As well as providing a resource for further research into the roles of microRNAs in immune responses in horses, this will facilitate inter-species comparative study of the role of microRNAs in the inflammatory cascade during endotoxemia and sepsis.


Asunto(s)
Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , MicroARNs/genética , Animales , Citocinas/biosíntesis , Secuenciación de Nucleótidos de Alto Rendimiento , Caballos , MicroARNs/sangre , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Genome Res ; 25(2): 226-34, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25367293

RESUMEN

V(D)J genomic recombination joins single gene segments to encode an extensive repertoire of antigen receptor specificities in T and B lymphocytes. This process initiates with double-stranded breaks adjacent to conserved recombination signal sequences that contain either 12- or 23-nucleotide spacer regions. Only recombination between signal sequences with unequal spacers results in productive coding genes, a phenomenon known as the "12/23 rule." Here we present two novel genomic tools that allow the capture and analysis of immune locus rearrangements from whole thymic and splenic tissues using second-generation sequencing. Further, we provide strong evidence that the 12/23 rule of genomic recombination is frequently violated under physiological conditions, resulting in unanticipated hybrid recombinations in ∼10% of Tcra excision circles. Hence, we demonstrate that strict adherence to the 12/23 rule is intrinsic neither to recombination signal sequences nor to the catalytic process of recombination and propose that nonclassical excision circles are liberated during the formation of antigen receptor diversity.


Asunto(s)
Genómica , Linfocitos/metabolismo , Recombinación V(D)J , Animales , Diferenciación Celular/genética , Reordenamiento Génico , Genes RAG-1 , Humanos , Linfocitos/citología , Ratones , Ratones Noqueados , Modelos Biológicos , Análisis de Secuencia de ADN
14.
Genome Res ; 22(1): 125-33, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22090378

RESUMEN

New sequencing technologies can address diverse biomedical questions but are limited by a minimum required DNA input of typically 1 µg. We describe how sequencing libraries can be reproducibly created from 20 pg of input DNA using a modified transpososome-mediated fragmentation technique. Resulting libraries incorporate in-line bar-coding, which facilitates sample multiplexes that can be sequenced using Illumina platforms with the manufacturer's sequencing primer. We demonstrate this technique by providing deep coverage sequence of the Escherichia coli K-12 genome that shows equivalent target coverage to a 1-µg input library prepared using standard Illumina methods. Reducing template quantity does, however, increase the proportion of duplicate reads and enriches coverage in low-GC regions. This finding was confirmed with exhaustive resequencing of a mouse library constructed from 20 pg of gDNA input (about seven haploid genomes) resulting in ∼0.4-fold statistical coverage of uniquely mapped fragments. This implies that a near-complete coverage of the mouse genome is obtainable with this approach using 20 genomes as input. Application of this new method now allows genomic studies from low mass samples and routine preparation of sequencing libraries from enrichment procedures.


Asunto(s)
ADN Bacteriano/química , Escherichia coli K12/química , Biblioteca de Genes , Análisis de Secuencia de ADN/métodos , Animales , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Escherichia coli K12/genética , Ratones
15.
J Neurosci ; 31(14): 5483-94, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21471385

RESUMEN

The cytoplasmic dynein complex is fundamentally important to all eukaryotic cells for transporting a variety of essential cargoes along microtubules within the cell. This complex also plays more specialized roles in neurons. The complex consists of 11 types of protein that interact with each other and with external adaptors, regulators and cargoes. Despite the importance of the cytoplasmic dynein complex, we know comparatively little of the roles of each component protein, and in mammals few mutants exist that allow us to explore the effects of defects in dynein-controlled processes in the context of the whole organism. Here we have taken a genotype-driven approach in mouse (Mus musculus) to analyze the role of one subunit, the dynein light intermediate chain 1 (Dync1li1). We find that, surprisingly, an N235Y point mutation in this protein results in altered neuronal development, as shown from in vivo studies in the developing cortex, and analyses of electrophysiological function. Moreover, mutant mice display increased anxiety, thus linking dynein functions to a behavioral phenotype in mammals for the first time. These results demonstrate the important role that dynein-controlled processes play in the correct development and function of the mammalian nervous system.


Asunto(s)
Conducta Animal/fisiología , Dineínas Citoplasmáticas/genética , Regulación del Desarrollo de la Expresión Génica/genética , Fenotipo , Mutación Puntual/genética , Animales , Animales Recién Nacidos , Asparagina/genética , Recuento de Células/métodos , Células Cultivadas , Corteza Cerebral/citología , Dendritas/genética , Embrión de Mamíferos , Femenino , Fibroblastos/fisiología , Fibroblastos/ultraestructura , Ganglios Espinales/citología , Proteínas Fluorescentes Verdes/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/genética , Proteínas del Tejido Nervioso , Conducción Nerviosa/genética , Neuronas/clasificación , Neuronas/citología , Neuronas/fisiología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Desempeño Psicomotor , Estadísticas no Paramétricas , Tirosina/genética , Levantamiento de Peso/fisiología
16.
PLoS Genet ; 5(12): e1000773, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20019802

RESUMEN

Spinal muscular atrophy is a severe motor neuron disease caused by inactivating mutations in the SMN1 gene leading to reduced levels of full-length functional SMN protein. SMN is a critical mediator of spliceosomal protein assembly, and complete loss or drastic reduction in protein leads to loss of cell viability. However, the reason for selective motor neuron degeneration when SMN is reduced to levels which are tolerated by all other cell types is not currently understood. Widespread splicing abnormalities have recently been reported at end-stage in a mouse model of SMA, leading to the proposition that disruption of efficient splicing is the primary mechanism of motor neuron death. However, it remains unclear whether splicing abnormalities are present during early stages of the disease, which would be a requirement for a direct role in disease pathogenesis. We performed exon-array analysis of RNA from SMN deficient mouse spinal cord at 3 time points, pre-symptomatic (P1), early symptomatic (P7), and late-symptomatic (P13). Compared to littermate control mice, SMA mice showed a time-dependent increase in the number of exons showing differential expression, with minimal differences between genotypes at P1 and P7, but substantial variation in late-symptomatic (P13) mice. Gene ontology analysis revealed differences in pathways associated with neuronal development as well as cellular injury. Validation of selected targets by RT-PCR confirmed the array findings and was in keeping with a shift between physiologically occurring mRNA isoforms. We conclude that the majority of splicing changes occur late in SMA and may represent a secondary effect of cell injury, though we cannot rule out significant early changes in a small number of transcripts crucial to motor neuron survival.


Asunto(s)
Empalme Alternativo/genética , Atrofia Muscular Espinal/patología , Animales , Modelos Animales de Enfermedad , Exones , Regulación de la Expresión Génica , Ratones , Neuronas Motoras , Isoformas de Proteínas , ARN Mensajero/análisis , Médula Espinal , Factores de Tiempo
17.
Neurobiol Dis ; 34(3): 511-7, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19332122

RESUMEN

Mutations in the ubiquitously expressed survival motor neuron 1 (SMN1) and superoxide dismutase 1 (SOD1) genes are selectively lethal to motor neurons in spinal muscular atrophy (SMA) and familial amyotrophic lateral sclerosis (ALS), respectively. Genetic association studies provide compelling evidence that SMN1 and SMN2 genotypes encoding lower SMN protein levels are implicated in sporadic ALS, suggesting that SMN expression is a potential determinant of ALS severity. We therefore sought genetic evidence of SMN involvement in ALS by generating transgenic mutant SOD1 mice on an Smn deficient background. Partial genetic disruption of Smn significantly worsened motor performance and survival in transgenic SOD1(G93A) mice. Furthermore, ALS-linked mutant SOD1 expression severely reduced SMN protein levels, but not transcript, in neuronal culture and mouse models from early presymptomatic disease. SMN protein depletion was linked to the nuclear compartment and a physical interaction between SMN and mutant SOD1 was confirmed in mouse spinal cord. Treatment with the environmental toxin paraquat also depleted SMN protein, implicating oxidative stress in the mechanism underlying SMN deficiency in familial ALS and potentially sporadic disease. In contrast, transgenic SOD1(WT) overexpression in SMA type I mice was incapable of modulating SMN protein levels or disease progression. These data establish that SMN deficiency accelerates phenotypic severity in transgenic familial ALS mice, consistent with an enhancing genetic modifier role. We therefore propose that SMN replacement and upregulation strategies considered for SMA therapy may have protective potential for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Superóxido Dismutasa/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/mortalidad , Animales , Línea Celular , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación Missense , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidantes/farmacología , Paraquat/farmacología , ARN Mensajero/metabolismo , Médula Espinal/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo
18.
BMC Neurosci ; 9: 104, 2008 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-18957104

RESUMEN

BACKGROUND: Redistribution of nuclear TAR DNA binding protein 43 (TDP-43) to the cytoplasm and ubiquitinated inclusions of spinal motor neurons and glial cells is characteristic of amyotrophic lateral sclerosis (ALS) pathology. Recent evidence suggests that TDP-43 pathology is common to sporadic ALS and familial ALS without SOD1 mutation, but not SOD1-related fALS cases. Furthermore, it remains unclear whether TDP-43 abnormalities occur in non-ALS forms of motor neuron disease. Here, we characterise TDP-43 localisation, expression levels and post-translational modifications in mouse models of ALS and spinal muscular atrophy (SMA). RESULTS: TDP-43 mislocalisation to ubiquitinated inclusions or cytoplasm was notably lacking in anterior horn cells from transgenic mutant SOD1G93A mice. In addition, abnormally phosphorylated or truncated TDP-43 species were not detected in fractionated ALS mouse spinal cord or brain. Despite partial colocalisation of TDP-43 with SMN, depletion of SMN- and coilin-positive Cajal bodies in motor neurons of affected SMA mice did not alter nuclear TDP-43 distribution, expression or biochemistry in spinal cords. CONCLUSION: These results emphasise that TDP-43 pathology characteristic of human sporadic ALS is not a core component of the neurodegenerative mechanisms caused by SOD1 mutation or SMN deficiency in mouse models of ALS and SMA, respectively.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/metabolismo , Atrofia Muscular Espinal/metabolismo , Médula Espinal/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Células del Asta Anterior/metabolismo , Células del Asta Anterior/patología , Núcleo Celular/metabolismo , Núcleo Celular/patología , Cuerpos Enrollados/patología , Citoplasma/metabolismo , Citoplasma/patología , Modelos Animales de Enfermedad , Femenino , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Procesamiento Proteico-Postraduccional/genética , Transporte de Proteínas/genética , Médula Espinal/patología , Superóxido Dismutasa/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Ubiquitinación
20.
PLoS Genet ; 2(10): e149, 2006 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-17029558

RESUMEN

Otitis media (OM), inflammation of the middle ear, remains the most common cause of hearing impairment in children. It is also the most common cause of surgery in children in the developed world. There is evidence from studies of the human population and mouse models that there is a significant genetic component predisposing to OM, yet nothing is known about the underlying genetic pathways involved in humans. We identified an N-ethyl-N-nitrosourea-induced dominant mouse mutant Junbo with hearing loss due to chronic suppurative OM and otorrhea. This develops from acute OM that arises spontaneously in the postnatal period, with the age of onset and early severity dependent on the microbiological status of the mice and their air quality. We have identified the causal mutation, a missense change in the C-terminal zinc finger region of the transcription factor Evi1. This protein is expressed in middle ear basal epithelial cells, fibroblasts, and neutrophil leukocytes at postnatal day 13 and 21 when inflammatory changes are underway. The identification and characterization of the Junbo mutant elaborates a novel role for Evi1 in mammalian disease and implicates a new pathway in genetic predisposition to OM.


Asunto(s)
Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Otitis Media/genética , Proto-Oncogenes/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Clonación Molecular , Análisis Mutacional de ADN , Proteínas de Unión al ADN/química , Oído Medio/citología , Oído Medio/patología , Citometría de Flujo , Granulocitos/inmunología , Pulmón/citología , Pulmón/patología , Proteína del Locus del Complejo MDS1 y EV11 , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Datos de Secuencia Molecular , Nariz/citología , Nariz/patología , Otitis Media/inmunología , Fenotipo , Organismos Libres de Patógenos Específicos , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...