Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Ecol Lett ; 25(4): 876-888, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35092147

RESUMEN

Viruses and their hosts can undergo coevolutionary arms races where hosts evolve increased resistance and viruses evolve counter-resistance. Given these arms race dynamics (ARD), both players are predicted to evolve along a single trajectory as more recently evolved genotypes replace their predecessors. By coupling phenotypic and genomic analyses of coevolving populations of bacteriophage λ and Escherichia coli, we find conflicting evidence for ARD. Virus-host infection phenotypes fit the ARD model, yet genomic analyses revealed fluctuating selection dynamics. Rather than coevolution unfolding along a single trajectory, cryptic genetic variation emerges and is maintained at low frequency for generations until it eventually supplants dominant lineages. These observations suggest a hybrid 'leapfrog' dynamic, revealing weaknesses in the predictive power of standard coevolutionary models. The findings shed light on the mechanisms that structure coevolving ecological networks and reveal the limits of using phenotypic or genomic data alone to differentiate coevolutionary dynamics.


Asunto(s)
Bacteriófagos , Bacterias/genética , Bacteriófagos/genética , Evolución Biológica , Fenotipo , Secuenciación Completa del Genoma
2.
Mol Cell Neurosci ; 71: 13-24, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26658803

RESUMEN

Discrepancy in synaptic structural plasticity is one of the earliest manifestations of the neurodegenerative state. In prion diseases, a reduction in synapses and dendritic spine densities is observed during preclinical disease in neurons of the cortex and hippocampus. The underlying molecular mechanisms of these alterations have not been identified but microRNAs (miRNAs), many of which are enriched at the synapse, likely regulate local protein synthesis in rapid response to stressors such as replicating prions. MiRNAs are therefore candidate regulators of these early neurodegenerative changes and may provide clues as to the molecular pathways involved. We therefore determined changes in mature miRNA abundance within synaptoneurosomes isolated from prion-infected, as compared to mock-infected animals, at asymptomatic and symptomatic stages of disease. During preclinical disease, miRNAs that are enriched in neurons including miR-124a-3p, miR-136-5p and miR-376a-3p were elevated. At later stages of disease we found increases in miRNAs that have previously been identified as deregulated in brain tissues of prion infected mice, as well as in Alzheimer's disease (AD) models. These include miR-146a-5p, miR-142-3p, miR-143-3p, miR-145a-5p, miR-451a, miR-let-7b, miR-320 and miR-150-5p. A number of miRNAs also decreased in abundance during clinical disease. These included almost all members of the related miR-200 family (miR-200a-3p, miR-200b-3p, miR-200c-3p, miR-141-3p, and miR-429-3p) and the 182 cluster (miR-182-5p and miR-183-5p).


Asunto(s)
MicroARNs/genética , Enfermedades por Prión/metabolismo , Sinapsis/metabolismo , Animales , Dendritas/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Ratones , Priones/metabolismo
3.
Hum Mutat ; 35(10): 1233-48, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25074322

RESUMEN

The involvement of SNPs in miRNA target sites remains poorly investigated in neurodegenerative disease. In addition to associations with disease risk, such genetic variations can also provide novel insight into mechanistic pathways that may be responsible for disease etiology and/or pathobiology. To identify SNPs associated specifically with degenerating neurons, we restricted our analysis to genes that are dysregulated in CA1 hippocampal neurons of mice during early, preclinical phase of Prion disease. The 125 genes chosen are also implicated in other numerous degenerative and neurological diseases and disorders and are therefore likely to be of fundamental importance. We predicted those SNPs that could increase, decrease, or have neutral effects on miRNA binding. This group of genes was more likely to possess DNA variants than were genes chosen at random. Furthermore, many of the SNPs are common within the human population, and could contribute to the growing awareness that miRNAs and associated SNPs could account for detrimental neurological states. Interestingly, SNPs that overlapped miRNA-binding sites in the 3'-UTR of GABA-receptor subunit coding genes were particularly enriched. Moreover, we demonstrated that SNP rs9291296 would strengthen miR-26a-5p binding to a highly conserved site in the 3'-UTR of gamma-aminobutyric acid receptor subunit alpha-4.


Asunto(s)
Regiones no Traducidas 3' , Sitios de Unión/genética , Regulación de la Expresión Génica , MicroARNs , Enfermedades Neurodegenerativas/genética , Polimorfismo de Nucleótido Simple , Enfermedades por Prión/genética , Animales , Región CA1 Hipocampal/metabolismo , Simulación por Computador , Bases de Datos de Ácidos Nucleicos , Humanos , Ratones , Neuronas/metabolismo , Receptores de GABA/genética
4.
PLoS Pathog ; 8(11): e1003002, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144617

RESUMEN

Prion diseases typically have long pre-clinical incubation periods during which time the infectious prion particle and infectivity steadily propagate in the brain. Abnormal neuritic sprouting and synaptic deficits are apparent during pre-clinical disease, however, gross neuronal loss is not detected until the onset of the clinical phase. The molecular events that accompany early neuronal damage and ultimately conclude with neuronal death remain obscure. In this study, we used laser capture microdissection to isolate hippocampal CA1 neurons and determined their pre-clinical transcriptional response during infection. We found that gene expression within these neurons is dynamic and characterized by distinct phases of activity. We found that a major cluster of genes is altered during pre-clinical disease after which expression either returns to basal levels, or alternatively undergoes a direct reversal during clinical disease. Strikingly, we show that this cluster contains a signature highly reminiscent of synaptic N-methyl-D-aspartic acid (NMDA) receptor signaling and the activation of neuroprotective pathways. Additionally, genes involved in neuronal projection and dendrite development were also altered throughout the disease, culminating in a general decline of gene expression for synaptic proteins. Similarly, deregulated miRNAs such as miR-132-3p, miR-124a-3p, miR-16-5p, miR-26a-5p, miR-29a-3p and miR-140-5p follow concomitant patterns of expression. This is the first in depth genomic study describing the pre-clinical response of hippocampal neurons to early prion replication. Our findings suggest that prion replication results in the persistent stimulation of a programmed response that is mediated, at least in part, by synaptic NMDA receptor activity that initially promotes cell survival and neurite remodelling. However, this response is terminated prior to the onset of clinical symptoms in the infected hippocampus, seemingly pointing to a critical juncture in the disease. Manipulation of these early neuroprotective pathways may redress the balance between degeneration and survival, providing a potential inroad for treatment.


Asunto(s)
Regulación de la Expresión Génica , Hipocampo/metabolismo , MicroARNs/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/metabolismo , Enfermedades por Prión/metabolismo , Priones/metabolismo , Animales , Estudio de Asociación del Genoma Completo , Hipocampo/patología , Hipocampo/fisiopatología , Ratones , Neuronas/patología , Enfermedades por Prión/patología , Enfermedades por Prión/fisiopatología
5.
Nat Commun ; 15(1): 863, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38286804

RESUMEN

A major challenge in evolutionary biology is explaining how populations navigate rugged fitness landscapes without getting trapped on local optima. One idea illustrated by adaptive dynamics theory is that as populations adapt, their newly enhanced capacities to exploit resources alter fitness payoffs and restructure the landscape in ways that promote speciation by opening new adaptive pathways. While there have been indirect tests of this theory, to our knowledge none have measured how fitness landscapes deform during adaptation, or test whether these shifts promote diversification. Here, we achieve this by studying bacteriophage [Formula: see text], a virus that readily speciates into co-existing receptor specialists under controlled laboratory conditions. We use a high-throughput gene editing-phenotyping technology to measure [Formula: see text]'s fitness landscape in the presence of different evolved-[Formula: see text] competitors and find that the fitness effects of individual mutations, and their epistatic interactions, depend on the competitor. Using these empirical data, we simulate [Formula: see text]'s evolution on an unchanging landscape and one that recapitulates how the landscape deforms during evolution. [Formula: see text] heterogeneity only evolves in the shifting landscape regime. This study provides a test of adaptive dynamics, and, more broadly, shows how fitness landscapes dynamically change during adaptation, potentiating phenomena like speciation by opening new adaptive pathways.


Asunto(s)
Bacteriófago lambda , Aptitud Genética , Bacteriófago lambda/genética , Retroalimentación , Mutación , Modelos Genéticos , Evolución Biológica
6.
J Biol Chem ; 287(20): 16510-20, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22447932

RESUMEN

The ubiquitously expressed cellular prion protein (PrP(C)) is subjected to the physiological α-cleavage at a region critical for both PrP toxicity and the conversion of PrP(C) to its pathogenic prion form (PrP(Sc)), generating the C1 and N1 fragments. The C1 fragment can activate caspase 3 while the N1 fragment is neuroprotective. Recent articles indicate that ADAM10, ADAM17, and ADAM9 may not play a prominent role in the α-cleavage of PrP(C) as previously thought, raising questions on the identity of the responsible protease(s). Here we show that, ADAM8 can directly cleave PrP to generate C1 in vitro and PrP C1/full-length ratio is greatly decreased in the skeletal muscles of ADAM8 knock-out mice; in addition, the PrP C1/full-length ratio is linearly correlated with ADAM8 protein level in myoblast cell line C2C12 and in skeletal muscle tissues of transgenic mice. These results indicate that ADAM8 is the primary protease responsible for the α-cleavage of PrP(C) in muscle cells. Moreover, we found that overexpression of PrP(C) led to up-regulation of ADAM8, suggesting that PrP(C) may regulate its own α-cleavage through modulating ADAM8 activity.


Asunto(s)
Proteínas ADAM/metabolismo , Antígenos CD/metabolismo , Proteínas de la Membrana/metabolismo , Mioblastos Esqueléticos/metabolismo , Proteínas PrPC/metabolismo , Proteolisis , Proteínas ADAM/genética , Animales , Antígenos CD/genética , Línea Celular , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Especificidad de Órganos/genética , Proteínas PrPC/genética
7.
Prion ; 17(1): 116-132, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37131335

RESUMEN

Prions are misfolded proteins that accumulate within the brain in association with a rare group of fatal and infectious neurological disorders in humans and animals. A current challenge to research is a lack of in vitro model systems that are compatible with a wide range of prion strains, reproduce prion toxicity, and are amenable to genetic manipulations. In an attempt to address this need, here we produced stable cell lines that overexpress different versions of PrPC through lentiviral transduction of immortalized human neural progenitor cells (ReN VM). Differentiated cultures made from the neural progenitor cell lines overexpressed PrPC within 3D spheroid-like structures of TUBB3+ neurons and we observed evidence that PrPC modulates formation of these structures, consistent with PrPC's role in neurogenesis. However, through repeated measurements of amyloid seeding activity in 6-week time course experiments, we failed to observe any evidence of prion replication within the differentiated ReN cultures following challenge with four prion isolates (human sCJD subtypes MM1 and VV2, and rodent adapted scrapie strains RML and 263K). We attributed amyloid seeding activity detected within the cultures to residual inoculum and concluded that PrPC overexpression was insufficient to confer permissiveness of ReN cultures to prion infection. While our ReN cell prion infection model was unsuccessful, additional efforts to develop cellular models of human prion disease are highly warranted.


Asunto(s)
Síndrome de Creutzfeldt-Jakob , Enfermedades por Prión , Priones , Animales , Humanos , Priones/metabolismo , Síndrome de Creutzfeldt-Jakob/genética , Enfermedades por Prión/metabolismo , Línea Celular , Células Madre/metabolismo
8.
Hear Res ; 437: 108853, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37441879

RESUMEN

Bimodal hearing, in which a contralateral hearing aid is combined with a cochlear implant (CI), provides greater speech recognition benefits than using a CI alone. Factors predicting individual bimodal patient success are not fully understood. Previous studies have shown that bimodal benefits may be driven by a patient's ability to extract fundamental frequency (f0) and/or temporal fine structure cues (e.g., F1). Both of these features may be represented in frequency following responses (FFR) to bimodal speech. Thus, the goals of this study were to: 1) parametrically examine neural encoding of f0 and F1 in simulated bimodal speech conditions; 2) examine objective discrimination of FFRs to bimodal speech conditions using machine learning; 3) explore whether FFRs are predictive of perceptual bimodal benefit. Three vowels (/ε/, /i/, and /ʊ/) with identical f0 were manipulated by a vocoder (right ear) and low-pass filters (left ear) to create five bimodal simulations for evoking FFRs: Vocoder-only, Vocoder +125 Hz, Vocoder +250 Hz, Vocoder +500 Hz, and Vocoder +750 Hz. Perceptual performance on the BKB-SIN test was also measured using the same five configurations. Results suggested that neural representation of f0 and F1 FFR components were enhanced with increasing acoustic bandwidth in the simulated "non-implanted" ear. As spectral differences between vowels emerged in the FFRs with increased acoustic bandwidth, FFRs were more accurately classified and discriminated using a machine learning algorithm. Enhancement of f0 and F1 neural encoding with increasing bandwidth were collectively predictive of perceptual bimodal benefit on a speech-in-noise task. Given these results, FFR may be a useful tool to objectively assess individual variability in bimodal hearing.


Asunto(s)
Implantación Coclear , Implantes Cocleares , Audífonos , Percepción del Habla , Humanos , Habla , Percepción del Habla/fisiología
9.
bioRxiv ; 2023 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-37645887

RESUMEN

A major challenge in evolutionary biology is explaining how populations navigate rugged fitness landscapes without getting trapped on local optima. One idea illustrated by adaptive dynamics theory is that as populations adapt, their newly enhanced capacities to exploit resources alter fitness payoffs and restructure the landscape in ways that promote speciation by opening new adaptive pathways. While there have been indirect tests of this theory, none have measured how fitness landscapes deform during adaptation, or test whether these shifts promote diversification. Here, we achieve this by studying bacteriophage λ, a virus that readily speciates into co-existing receptor specialists under controlled laboratory conditions. We used a high-throughput gene editing-phenotyping technology to measure λ's fitness landscape in the presence of different evolved-λ competitors and found that the fitness effects of individual mutations, and their epistatic interactions, depend on the competitor. Using these empirical data, we simulated λ's evolution on an unchanging landscape and one that recapitulates how the landscape deforms during evolution. λ heterogeneity only evolved in the shifting landscape regime. This study provides a test of adaptive dynamics, and, more broadly, shows how fitness landscapes dynamically change during adaptation, potentiating phenomena like speciation by opening new adaptive pathways.

10.
Acta Neuropathol Commun ; 10(1): 161, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36352465

RESUMEN

Prion diseases are neurodegenerative disorders with long asymptomatic incubation periods, followed by a rapid progression of cognitive and functional decline culminating in death. The complexity of intercellular interactions in the brain is challenging to unravel and the basis of disease pathobiology remains poorly understood. In this study, we employed single cell RNA sequencing (scRNAseq) to produce an atlas of 147,536 single cell transcriptomes from cortex and hippocampus of mice infected with prions and showing clinical signs. We identified transcriptionally distinct populations and sub-populations of all the major brain cell-types. Disease-related transcription was highly specific to not only overarching cell-types, but also to sub-populations of glia and neurons. Most striking was an apparent decrease in relative frequency of astrocytes expressing genes that are required for brain homeostasis such as lipid synthesis, glutamate clearance, synaptic modulation and regulation of blood flow. Additionally, we described a spectrum of microglial activation states that suggest delineation of phagocytic and neuroinflammatory functions in different cell subsets. Differential responses of immature and mature neuron populations were also observed, alongside abnormal hippocampal neurogenesis. Our scRNAseq library provides a new layer of knowledge on single cell gene expression in prion disease, and is a basis for a more detailed understanding of cellular interplay that leads to neurodegeneration.


Asunto(s)
Astrocitos , Enfermedades por Prión , Animales , Ratones , Astrocitos/metabolismo , Microglía/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Hipocampo/metabolismo , Neurogénesis , Análisis de Secuencia de ARN
11.
Front Neurosci ; 16: 918811, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35651626

RESUMEN

Progressive dysfunction and loss of neurons ultimately culminates in the symptoms and eventual fatality of prion disease, yet the pathways and mechanisms that lead to neuronal degeneration remain elusive. Here, we used RNAseq to profile transcriptional changes in microdissected CA1 and thalamus brain tissues from prion infected mice. Numerous transcripts were altered during clinical disease, whereas very few transcripts were reliably altered at pre-clinical time points. Prion altered transcripts were assigned to broadly defined brain cell types and we noted a strong transcriptional signature that was affiliated with reactive microglia and astrocytes. While very few neuronal transcripts were common between the CA1 and thalamus, we described transcriptional changes in both regions that were related to synaptic dysfunction. Using transcriptional profiling to compare how different neuronal populations respond during prion disease may help decipher mechanisms that lead to neuronal demise and should be investigated with greater detail.

12.
Viruses ; 14(6)2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35746689

RESUMEN

The numerous neurological syndromes associated with COVID-19 implicate an effect of viral pathogenesis on neuronal function, yet reports of direct SARS-CoV-2 infection in the brain are conflicting. We used a well-established organotypic brain slice culture to determine the permissivity of hamster brain tissues to SARS-CoV-2 infection. We found levels of live virus waned after inoculation and observed no evidence of cell-to-cell spread, indicating that SARS-CoV-2 infection was non-productive. Nonetheless, we identified a small number of infected cells with glial phenotypes; however, no evidence of viral infection or replication was observed in neurons. Our data corroborate several clinical studies that have assessed patients with COVID-19 and their association with neurological involvement.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Encéfalo , Cricetinae , Humanos , Neuroglía , Neuronas
13.
J Toxicol Environ Health A ; 74(22-24): 1504-20, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22043911

RESUMEN

Prion replication in the periphery precedes neuroinvasion in many experimental rodent scrapie models, and in natural sheep scrapie and chronic wasting disease (CWD) in cervids. Prions propagate in the germinal centers of secondary lymphoid organs and are strongly associated with follicular dendritic cells (FDC) and possibly circulating dendritic cells and macrophages. Given the importance of lymphoid organs in prion disease transmission and pathogenesis, gene expression studies may reveal host factors or biological pathways related to prion replication and accumulation. A procedure was developed to enrich for FDC, dendritic cells, and macrophages prior to the investigation of transcriptional alterations in murine splenic cells during prion pathogenesis. In total, 1753 transcripts exhibited fold changes greater than three (false discovery rates less than 2%) in this population isolated from spleens of prion-infected versus uninfected mice. The gene for the small leucine-rich proteoglycan decorin (DCN) was one of the genes most overexpressed in infected mice, and the splenic protein levels mirrored this in mice infected with scrapie as well as bovine spongiform encephalopathy (BSE) and variant Creutzfeldt-Jakob disease (vCJD). A number of groups of functionally related genes were also significantly decreased in infected spleens. These included genes related to iron metabolism and homeostasis, pathways that have also been implicated in prion pathogenesis in the brain. These gene expression alterations provide insights into the molecular mechanisms underlying prion disease pathogenesis and may serve as a pool of potential surrogate markers for the early detection and diagnosis of some prion diseases.


Asunto(s)
Decorina , Leucocitos/inmunología , Enfermedades por Prión/diagnóstico , Priones/inmunología , Bazo/inmunología , Transcripción Genética , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Bovinos , Decorina/genética , Decorina/inmunología , Decorina/metabolismo , Células Dendríticas Foliculares/citología , Células Dendríticas Foliculares/inmunología , Células Dendríticas Foliculares/metabolismo , Femenino , Leucocitos/citología , Leucocitos/metabolismo , Ratones , Enfermedades por Prión/inmunología , Enfermedades por Prión/transmisión , Enfermedades por Prión/veterinaria , Priones/química , Priones/genética , Priones/metabolismo , Bazo/citología , Bazo/metabolismo
14.
Evolution ; 74(4): 764-774, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31891185

RESUMEN

An important driver of evolution in viruses is natural selection to optimize the use of their hosts' genetic network. To learn how viruses respond to this pressure, we disrupted the genetic network of Escherichia coli to inhibit replication of its virus, bacteriophage lambda, and then observed how λ evolved to compensate. We deleted E. coli's dnaJ gene, which lambda uses to initiate DNA replication. Lambda partially restored its ability to reproduce with just two adaptive mutations associated with genes J and S. The location of the mutations was unexpected because they were not in genes that directly interact with DnaJ, rather they affected seemingly unrelated life history traits. A nonsynonymous J mutation increased lambda's adsorption rate and an S regulatory mutation delayed lysis timing. Lambda also recovered some of its reproductive potential through intracellular mutualism. This study offers two important lessons: first, viruses can rapidly adapt to disruptive changes in their host's genetic network. Second, organisms can employ mechanisms thought to operate at the population scale, such as evolution of life history traits and social interactions, in order to overcome hurdles at the molecular level. As life science research progresses and new fields become increasingly specialized, these results remind us of the importance of multiscale and interdisciplinary approaches to understand adaptation.


Asunto(s)
Bacteriófago lambda/fisiología , Evolución Biológica , Escherichia coli/virología , Interacciones Microbiota-Huesped , Rasgos de la Historia de Vida , Simbiosis , Bacteriófago lambda/genética , Genes Virales , Mutación , Simbiosis/genética
15.
Mol Oncol ; 14(3): 520-538, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31930767

RESUMEN

Breast cancer brain metastases (BCBMs) have been underinvestigated despite their high incidence and poor outcome. MicroRNAs (miRNAs), and particularly circulating miRNAs, regulate multiple cellular functions, and their deregulation has been reported in different types of cancer and metastasis. However, their signature in plasma along brain metastasis development and their relevant targets remain undetermined. Here, we used a mouse model of BCBM and next-generation sequencing (NGS) to establish the alterations in circulating miRNAs during brain metastasis formation and development. We further performed bioinformatics analysis to identify their targets with relevance in the metastatic process. We additionally analyzed human resected brain metastasis samples of breast cancer patients for target expression validation. Breast cancer cells were injected in the carotid artery of mice to preferentially induce metastasis in the brain, and samples were collected at different timepoints (5 h, 3, 7, and 10 days) to follow metastasis development in the brain and in peripheral organs. Metastases were detected from 7 days onwards, mainly in the brain. NGS revealed a deregulation of circulating miRNA profile during BCBM progression, rising from 18% at 3 days to 30% at 10 days following malignant cells' injection. Work was focused on those altered prior to metastasis detection, among which were miR-802-5p and miR-194-5p, whose downregulation was validated by qPCR. Using targetscan and diana tools, the transcription factor myocyte enhancer factor 2C (MEF2C) was identified as a target for both miRNAs, and its expression was increasingly observed in malignant cells along brain metastasis development. Its upregulation was also observed in peritumoral astrocytes pointing to a role of MEF2C in the crosstalk between tumor cells and astrocytes. MEF2C expression was also observed in human BCBM, validating the observation in mouse. Collectively, downregulation of circulating miR-802-5p and miR-194-5p appears as a precocious event in BCBM and MEF2C emerges as a new player in brain metastasis development.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias Mamarias Animales/sangre , MicroARNs/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Biología Computacional , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/secundario , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
16.
PLoS Negl Trop Dis ; 14(4): e0008105, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32251473

RESUMEN

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus that can cause a hemorrhagic fever in humans, with a case fatality rate of up to 40%. Cases of CCHFV have been reported in Africa, Asia, and southern Europe; and recently, due to the expanding range of its vector, autochthonous cases have been reported in Spain. Although it was discovered over 70 years ago, our understanding of the pathogenesis of this virus remains limited. We used RNA-Seq in two human liver cell lines (HepG2 and Huh7) infected with CCHFV (strain IbAr10200), to examine kinetic changes in host expression and viral replication simultaneously at 1 and 3 days post infection. Through this, numerous host pathways were identified that were modulated by the virus including: antiviral response and endothelial cell leakage. Notably, the genes encoding DDX60, a cytosolic component of the RIG-I signalling pathway and OAS2 were both shown to be dysregulated. Interestingly, PTPRR was induced in Huh7 cells but not HepG2 cells. This has been associated with the TLR9 signalling cascade, and polymorphisms in TLR9 have been associated with poor outcomes in patients. Additionally, we performed whole-genome sequencing on CCHFV to assess viral diversity over time, and its relationship to the host response. As a result, we have demonstrated that through next-generation mRNA deep-sequencing it is possible to not only examine mRNA gene expression, but also to examine viral quasispecies and typing of the infecting strain. This demonstrates a proof-of-principle that CCHFV specimens can be analyzed to identify both the virus and host biomarkers that may have implications for prognosis.


Asunto(s)
Expresión Génica , Virus de la Fiebre Hemorrágica de Crimea-Congo/genética , Fiebre Hemorrágica de Crimea/genética , Interacciones Huésped-Patógeno/genética , Hígado/metabolismo , RNA-Seq/métodos , 2',5'-Oligoadenilato Sintetasa/genética , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Redes Reguladoras de Genes , Fiebre Hemorrágica de Crimea/metabolismo , Fiebre Hemorrágica de Crimea/virología , Células Hep G2 , Interacciones Huésped-Patógeno/fisiología , Humanos , ARN Mensajero , Receptores Inmunológicos , Transducción de Señal , Receptor Toll-Like 9 , Replicación Viral , Secuenciación del Exoma
17.
BMC Genomics ; 10: 201, 2009 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-19400950

RESUMEN

BACKGROUND: We have reported that doxycycline-induced over-expression of wild type prion protein (PrP) in skeletal muscles of Tg(HQK) mice is sufficient to cause a primary myopathy with no signs of peripheral neuropathy. The preferential accumulation of the truncated PrP C1 fragment was closely correlated with these myopathic changes. In this study we use gene expression profiling to explore the temporal program of molecular changes underlying the PrP-mediated myopathy. RESULTS: We used DNA microarrays, and confirmatory real-time PCR and Western blot analysis to demonstrate deregulation of a large number of genes in the course of the progressive myopathy in the skeletal muscles of doxycycline-treated Tg(HQK) mice. These include the down-regulation of genes coding for the myofibrillar proteins and transcription factor MEF2c, and up-regulation of genes for lysosomal proteins that is concomitant with increased lysosomal activity in the skeletal muscles. Significantly, there was prominent up-regulation of p53 and p53-regulated genes involved in cell cycle arrest and promotion of apoptosis that paralleled the initiation and progression of the muscle pathology. CONCLUSION: The data provides the first in vivo evidence that directly links p53 to a wild type PrP-mediated disease. It is evident that several mechanistic features contribute to the myopathy observed in PrP over-expressing mice and that p53-related apoptotic pathways appear to play a major role.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Perfilación de la Expresión Génica , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Proteínas PrPC/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Doxiciclina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Enfermedades Musculares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Proteínas PrPC/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
J Toxicol Environ Health A ; 72(17-18): 1075-82, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19697242

RESUMEN

Prion and Alzheimer's diseases are two apparently distinct disorders; however, the two proteinaceous species implicated in disease progression share a number of common features. In prion diseases a beta-rich conformer of the prion protein is the key molecule in the pathogenesis of prion disease, whereas in Alzheimer's disease neurotoxicity is associated with the amyloid-beta peptide. These two molecules share common structural features and post-translational processing events and both undergo structural transition from normal host proteins to a form associated with toxicity, which leads to neurodegeneration. The precise mechanisms leading to neuronal damage and death that are triggered in these diseases are as yet unknown. It is possible, however, that there is a convergence of events in the neurons whereby similar pathways are executed. In this study the expression of a panel of 94 genes associated with the development of Alzheimer's disease was examined using a high-throughput real-time quantitative reverse-transcription polymerase chain reaction (RT-PCR) assay. Data showed that approximately 31 of these genes are deregulated in the brains of scrapie-infected mice. Among these were genes involved in inflammation, post-translational processing, excitotoxicity, cholesterol metabolism, and neuroprotection. One of the genes showing the greatest degree of upregulation was the cell cycle regulator CDC2. A microarray analysis also revealed deregulation of CDC2 and related genes, including cyclin B and cyclin D, suggesting that in prion disease, as in Alzheimer's disease, misregulation of cell cycle regulators may contribute to neurodegeneration.


Asunto(s)
Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Scrapie/genética , Animales , Encéfalo/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones
19.
Sci Rep ; 9(1): 1099, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30705335

RESUMEN

Multiple cell types and complex connection networks are an intrinsic feature of brain tissue. In this study we used expression profiling of specific microscopic regions of heterogeneous tissue sections isolated by laser capture microdissection (LCM) to determine insights into the molecular basis of brain pathology in prion disease. Temporal profiles in two mouse models of prion disease, bovine spongiform encephalopathy (BSE) and a mouse-adapted strain of scrapie (RML) were performed in microdissected regions of the CA1 hippocampus and granular layer of the cerebellum which are both enriched in neuronal cell bodies. We noted that during clinical disease the number of activated microglia and astrocytes that occur in these areas are increased, thereby likely diluting the neuronal gene expression signature. We performed a comparative analysis with gene expression profiles determined from isolated populations of neurons, microglia and astrocytes to identify transcripts that are enriched in each of these cell types. Although the incubation periods of these two models are quite different, over 300 days for BSE and ~160 days for RML scrapie, these regional microdissections revealed broadly similar profiles. Microglial and astrocyte-enriched genes contributed a profound inflammatory profile consisting of inflammatory cytokines, genes related to phagocytosis, proteolysis and genes coding for extracellular matrix proteins. CA1 pyramidal neurons displayed a net upregulation of transcription factors and stress induced genes at pre-clinical stages of disease while all tissues showed profound decrease of overlapping genes related to neuronal function, in particular transcripts related to neuronal communication including glutamate receptors, phosphatase subunits and numerous synapse-related markers. Of note, we found a small number of genes expressed in neurons that were upregulated during clinical disease including, COX6A2, FZD9, RXRG and SOX11, that may be biomarkers of neurodegeneration.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Cerebelo/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Enfermedades por Prión/metabolismo , Transcriptoma/genética , Animales , Ratones
20.
Sci Rep ; 9(1): 19705, 2019 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-31873177

RESUMEN

Chronic wasting disease (CWD) is an emerging infectious prion disorder that is spreading rapidly in wild populations of cervids in North America. The risk of zoonotic transmission of CWD is as yet unclear but a high priority must be to minimize further spread of the disease. No simple diagnostic tests are available to detect CWD quickly or in live animals; therefore, easily accessible biomarkers may be useful in identifying infected animals. MicroRNAs (miRNAs) are a class of small, non-coding RNA molecules that circulate in blood and are promising biomarkers for several infectious diseases. In this study we used next-generation sequencing to characterize the serum miRNA profiles of 35 naturally infected elk that tested positive for CWD in addition to 35 elk that tested negative for CWD. A total of 21 miRNAs that are highly conserved amongst mammals were altered in abundance in sera, irrespective of hemolysis in the samples. A number of these miRNAs have previously been associated with prion diseases. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the discriminative potential of these miRNAs as biomarkers for the diagnosis of CWD. We also determined that a subgroup of 6 of these miRNAs were consistently altered in abundance in serum from hamsters experimentally infected with scrapie. This suggests that common miRNA candidate biomarkers could be selected for prion diseases in multiple species. Furthermore, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses pointed to a strong correlation for 3 of these miRNAs, miR-148a-3p, miR-186-5p, miR-30e-3p, with prion disease.


Asunto(s)
MicroARN Circulante/sangre , MicroARN Circulante/genética , Ciervos/sangre , Ciervos/genética , Perfilación de la Expresión Génica , Enfermedad Debilitante Crónica/sangre , Enfermedad Debilitante Crónica/genética , Animales , Biomarcadores/sangre , Cricetinae/sangre , Cricetinae/genética , Redes Reguladoras de Genes , Anotación de Secuencia Molecular , Priones/metabolismo , Enfermedad Debilitante Crónica/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA