Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 34(10): 1319-35, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25759216

RESUMEN

Snail family members regulate epithelial-to-mesenchymal transition (EMT) during invasion of intestinal tumours, but their role in normal intestinal homeostasis is unknown. Studies in breast and skin epithelia indicate that Snail proteins promote an undifferentiated state. Here, we demonstrate that conditional knockout of Snai1 in the intestinal epithelium results in apoptotic loss of crypt base columnar stem cells and bias towards differentiation of secretory lineages. In vitro organoid cultures derived from Snai1 conditional knockout mice also undergo apoptosis when Snai1 is deleted. Conversely, ectopic expression of Snai1 in the intestinal epithelium in vivo results in the expansion of the crypt base columnar cell pool and a decrease in secretory enteroendocrine and Paneth cells. Following conditional deletion of Snai1, the intestinal epithelium fails to produce a proliferative response following radiation-induced damage indicating a fundamental requirement for Snai1 in epithelial regeneration. These results demonstrate that Snai1 is required for regulation of lineage choice, maintenance of CBC stem cells and regeneration of the intestinal epithelium following damage.


Asunto(s)
Mucosa Intestinal/metabolismo , Intestinos/citología , Factores de Transcripción/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Linaje de la Célula , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
2.
Neurochem Res ; 44(6): 1410-1424, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30661228

RESUMEN

Traumatic brain injury triggers neuroinflammation that may contribute to progressive neurodegeneration. We investigated patterns of recruitment of astrocytes and microglia to inflammation after brain trauma by firstly characterising expression profiles over time of marker genes following TBI, and secondly by monitoring glial morphologies reflecting inflammatory responses in a rat model of traumatic brain injury (i.e. the lateral fluid percussion injury). Gene expression profiles revealed early elevation of expression of astrocytic marker glial fibrillary acidic protein relative to microglial marker allograft inflammatory factor 1 (also known as ionized calcium-binding adapter molecule 1). Adult rat brains collected at day 7 after injury were processed for immunohistochemistry with allograft inflammatory factor 1, glial fibrillary acidic protein and complement C3 (marker of bad/disruptive astrocytic A1 phenotype). Astrocytes positive for glial fibrillary acidic protein and complement C3 were significant increased in the injured cortex and displayed more complex patterns of arbourisation with significantly increased bifurcations. Our observations suggested that traumatic brain injury changed the phenotype of microglia from a ramified appearance with long, thin, highly branched processes to a swollen amoeboid shape in the injured cortex. These findings suggest differential glial activation with astrocytes likely undergoing strategic changes in morphology and function. Whilst a detailed analysis is needed of temporal patterns of glial activation, ours is the first evidence of a role for the bad/disruptive astrocytic A1 phenotype in an open head model of traumatic brain injury.


Asunto(s)
Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Animales , Astrocitos/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Complemento C3/metabolismo , Equidae , Proteína Ácida Fibrilar de la Glía/metabolismo , Cabras , Masculino , Ratones , Microglía/patología , Conejos , Ratas Sprague-Dawley
3.
PLoS Biol ; 11(8): e1001625, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23966833

RESUMEN

The myelination of axons is a crucial step during vertebrate central nervous system (CNS) development, allowing for rapid and energy efficient saltatory conduction of nerve impulses. Accordingly, the differentiation of oligodendrocytes, the myelinating cells of the CNS, and their expression of myelin genes are under tight transcriptional control. We previously identified a putative transcription factor, Myelin Regulatory Factor (Myrf), as being vital for CNS myelination. Myrf is required for the generation of CNS myelination during development and also for its maintenance in the adult. It has been controversial, however, whether Myrf directly regulates transcription, with reports of a transmembrane domain and lack of nuclear localization. Here we show that Myrf is a membrane-associated transcription factor that undergoes an activating proteolytic cleavage to separate its transmembrane domain-containing C-terminal region from a nuclear-targeted N-terminal region. Unexpectedly, this cleavage event occurs via a protein domain related to the autoproteolytic intramolecular chaperone domain of the bacteriophage tail spike proteins, the first time this domain has been found to play a role in eukaryotic proteins. Using ChIP-Seq we show that the N-terminal cleavage product directly binds the enhancer regions of oligodendrocyte-specific and myelin genes. This binding occurs via a defined DNA-binding consensus sequence and strongly promotes the expression of target genes. These findings identify Myrf as a novel example of a membrane-associated transcription factor and provide a direct molecular mechanism for its regulation of oligodendrocyte differentiation and CNS myelination.


Asunto(s)
Proteínas de la Membrana/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Humanos , Proteínas de la Membrana/genética , Ratones , Mutagénesis Sitio-Dirigida , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Factores de Transcripción/genética
4.
Glia ; 63(6): 1005-20, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25639936

RESUMEN

In order to further investigate the molecular mechanisms that regulate oligodendrocyte (OC) survival, we utilized microarrays to characterize changes in OC gene expression after exposure to the cytokines neurotrophin3, insulin, or leukemia inhibitory factor (LIF) in vitro. We identified and validated the induction and secretion of the neuropeptide galanin in OCs, specifically in response to LIF. We next established that galanin is an OC survival factor and showed that autocrine or paracrine galanin secretion mediates LIF-induced OC survival in vitro. We also revealed that galanin is up-regulated in OCs in the cuprizone model of central demyelination, and that oligodendroglial galanin expression is significantly regulated by endogenous LIF in this context. We also showed that knock-out of galanin reduces OC survival and exacerbates callosal demyelination in the cuprizone model. These findings suggest a potential role for the use of galanin agonists in the treatment of human demyelinating diseases.


Asunto(s)
Galanina/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Animales , Astrocitos/patología , Astrocitos/fisiología , Encéfalo/patología , Encéfalo/fisiopatología , Supervivencia Celular/fisiología , Células Cultivadas , Cuprizona , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Galanina/genética , Expresión Génica , Sistema de Señalización de MAP Quinasas/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/patología , Células-Madre Neurales/patología , Células-Madre Neurales/fisiología , Oligodendroglía/patología , Nervio Óptico/patología , Nervio Óptico/fisiología , ARN Mensajero/metabolismo , Ratas Sprague-Dawley
5.
J Neurosci Res ; 92(6): 732-42, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24615902

RESUMEN

We conducted a microarray study to identify genes that are differentially regulated in the spinal cords of mice with the inflammatory disease experimental autoimmune encephalomyelitis (EAE) relative to healthy mice. In total 181 genes with at least a two-fold increase in expression were identified, and most of these genes were associated with immune function. Unexpectedly, ceruloplasmin (Cp), a ferroxidase that converts toxic ferrous iron to its nontoxic ferric form and also promotes the efflux of iron from astrocytes in the CNS, was shown to be highly upregulated (13.2-fold increase) in EAE spinal cord. Expression of Cp protein is known to be increased in several neurological conditions, but the role of Cp regulation in CNS autoimmune disease is not known. To investigate this, we induced EAE in Cp gene knockout, heterozygous, and wild-type mice. Cp knockout mice were found to have slower disease evolution than wild-type mice (EAE days 13-17; P = 0.05). Interestingly, Cp knockout mice also exhibited a significant increase in the number of astrocytes with reactive morphology in early EAE compared with wild-type mice at the same stage of disease. CNS iron levels were not increased with EAE in these mice. Based on these observations, we propose that an increase in Cp expression could contribute to tissue damage in early EAE. In addition, endogenous CP either directly or indirectly inhibits astrocyte reactivity during early disease, which could also worsen early disease evolution.


Asunto(s)
Ceruloplasmina/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Animales , Western Blotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/patología , Transcriptoma
6.
J Cell Physiol ; 227(3): 1199-211, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21604263

RESUMEN

Inhibitors of Rho kinase (ROCK) have potential for management of neurological disorders by inhibition of glial scarring. Since astrocytes play key roles in brain physiology and pathology, we determined changes in the astrocytic transcriptome produced by the ROCK inhibitor Fasudil to obtain mechanistic insights into its beneficial action during brain injury. Cultured murine astrocytes were treated with Fasudil (100 µM) and morphological analyses revealed rapid stellation by 1 h and time-dependent (2-24 h) dissipation of F-actin-labelled stress fibres. Microarray analyses were performed on RNA and the time-course of global gene profiling (2, 6, 12 and 24 h) provided a comprehensive description of transcriptomic changes. Hierarchical clustering of differentially expressed genes and analysis for over-represented gene ontology groups using the DAVID database focused attention on Fasudil-induced changes to major biological processes regulating cellular shape and motility (actin cytoskeleton, axon guidance, transforming growth factor-ß (TGFß) signalling and tight junctions). Bioinformatic analyses of transcriptomic changes revealed how these biological processes contributed to changes in astrocytic motility and cytoskeletal reorganisation. Here genes associated with extracellular matrix were also involved, but unexpected was a subset of alterations (EAAT2, BDNF, anti-oxidant species, metabolic and signalling genes) indicative of adoption by astrocytes of a pro-survival phenotype. Expression profiles of key changes with Fasudil and another ROCK inhibitor Y27632 were validated by real-time PCR. Although effects of ROCK inhibition have been considered to be primarily cytoskeletal via reduction of glial scarring, we demonstrate additional advantageous actions likely to contribute to their ameliorative actions in brain injury.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Perfilación de la Expresión Génica/métodos , Transcriptoma/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Astrocitos/citología , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Transcriptoma/genética , Quinasas Asociadas a rho/genética
7.
Ann Neurol ; 70(6): 897-912, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22190364

RESUMEN

OBJECTIVE: To perform a 1-stage meta-analysis of genome-wide association studies (GWAS) of multiple sclerosis (MS) susceptibility and to explore functional consequences of new susceptibility loci. METHODS: We synthesized 7 MS GWAS. Each data set was imputed using HapMap phase II, and a per single nucleotide polymorphism (SNP) meta-analysis was performed across the 7 data sets. We explored RNA expression data using a quantitative trait analysis in peripheral blood mononuclear cells (PBMCs) of 228 subjects with demyelinating disease. RESULTS: We meta-analyzed 2,529,394 unique SNPs in 5,545 cases and 12,153 controls. We identified 3 novel susceptibility alleles: rs170934(T) at 3p24.1 (odds ratio [OR], 1.17; p = 1.6 × 10(-8)) near EOMES, rs2150702(G) in the second intron of MLANA on chromosome 9p24.1 (OR, 1.16; p = 3.3 × 10(-8)), and rs6718520(A) in an intergenic region on chromosome 2p21, with THADA as the nearest flanking gene (OR, 1.17; p = 3.4 × 10(-8)). The 3 new loci do not have a strong cis effect on RNA expression in PBMCs. Ten other susceptibility loci had a suggestive p < 1 × 10(-6) , some of these loci have evidence of association in other inflammatory diseases (ie, IL12B, TAGAP, PLEK, and ZMIZ1). INTERPRETATION: We have performed a meta-analysis of GWAS in MS that more than doubles the size of previous gene discovery efforts and highlights 3 novel MS susceptibility loci. These and additional loci with suggestive evidence of association are excellent candidates for further investigations to refine and validate their role in the genetic architecture of MS.


Asunto(s)
Susceptibilidad a Enfermedades , Predisposición Genética a la Enfermedad , Esclerosis Múltiple/genética , Polimorfismo de Nucleótido Simple/genética , Adolescente , Adulto , Niño , Femenino , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Metaanálisis como Asunto , Persona de Mediana Edad , Esclerosis Múltiple/etiología , Oportunidad Relativa , Adulto Joven
8.
J Hepatol ; 55(3): 536-544, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21266183

RESUMEN

BACKGROUND & AIMS: Studies have shown that GB virus C (GBV-C) infection leads to reduced liver disease in hepatitis C virus (HCV)/human immunodeficiency virus (HIV) co-infection. Considering that the underlying mechanism(s) are unknown, we aim to identify differential gene and protein expression associated with GBV-C in HCV/HIV co-infection that may be responsible for reduced liver disease. METHODS: Liver, peripheral blood mononuclear cells (PBMCs), and plasma samples were collected from 43 HCV/HIV patients. Plasma was tested for GBV-C RNA by RT-PCR with NS5B gene primers. A microarray was performed on the liver and RT-qPCRs on the liver/PBMC samples. Hepatic protein expression was measured by immunohistochemistry. RESULTS: Sixteen out of 43 patients had GBV-C RNA. GBV-C was associated with reduced hepatic fibrosis (p=0.005) and inflammation (p=0.007). The microarray analysis of the liver samples (n=10) showed down-regulation of genes critical to intra-hepatic T-cell signaling associated with GBV-C. Quantitative RT-PCR of the liver samples (n=13) confirmed the down-regulation of lymphocyte-specific protein tyrosine kinase (LCK) (p=0.02) and docking protein 2 (DOK2) (p=0.04). No differences in the expression levels of these genes were observed in PBMCs (n=22) according to the GBV-C status. The hepatic expression of the LCK protein, measured by immunohistochemistry (n=36), was decreased in CD3-positive T-cells within portal tracts associated with GBV-C (p=0.003). This remained significant in multivariate analysis controlling for hepatic fibrosis and inflammation (p=0.027). No differences were observed in plasma cytokine concentrations (n=25) or ex-vivo peripheral T-cell responses (n=13) versus GBV-C status. CONCLUSIONS: GBV-C infection is associated with down-regulation of critical genes involved in intra-hepatic T-cell signaling in HCV/HIV co-infection. This may be relevant to the pathogenesis of reduced HCV-related liver disease in HIV co-infection.


Asunto(s)
Regulación hacia Abajo/genética , Infecciones por Flaviviridae/genética , Infecciones por Flaviviridae/metabolismo , Virus GB-C/metabolismo , Infecciones por VIH/complicaciones , Hepatitis Viral Humana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Complejo CD3/genética , Complejo CD3/metabolismo , Coinfección , Citocinas/sangre , Regulación hacia Abajo/inmunología , Femenino , Infecciones por Flaviviridae/sangre , Infecciones por Flaviviridae/complicaciones , Virus GB-C/inmunología , Infecciones por VIH/inmunología , Hepatitis C/complicaciones , Hepatitis C/inmunología , Hepatitis C/metabolismo , Hepatitis C/patología , Hepatitis Viral Humana/sangre , Hepatitis Viral Humana/complicaciones , Hepatitis Viral Humana/genética , Humanos , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , ARN Viral/sangre , Índice de Severidad de la Enfermedad , Transducción de Señal/genética , Transducción de Señal/inmunología , Células TH1/metabolismo , Células Th2/metabolismo
9.
Proteomics ; 10(12): 2377-95, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20391539

RESUMEN

The primary constituent of the amyloid plaque, beta-amyloid (Abeta), is thought to be the causal "toxic moiety" of Alzheimer's disease. However, despite much work focused on both Abeta and its parent protein, amyloid precursor protein (APP), the functional roles of APP and its cleavage products remain to be fully elucidated. Protein-protein interaction networks can provide insight into protein function, however, high-throughput data often report false positives and are in frequent disagreement with low-throughput experiments. Moreover, the complexity of the CNS is likely to be under represented in such databases. Therefore, we curated the published work characterizing both APP and Abeta to create a protein interaction network of APP and its proteolytic cleavage products, with annotation, where possible, to the level of APP binding domain and isoform. This is the first time that an interactome has been refined to domain level, essential for the interpretation of APP due to the presence of multiple isoforms and processed fragments. Gene ontology and network analysis were used to identify potentially novel functional relationships among interacting proteins.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Biología de Sistemas , Humanos , Unión Proteica
10.
J Neurosci ; 29(15): 4697-707, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19369540

RESUMEN

Using a novel microfluidic chamber that allows the isolation of axons without contamination by nonaxonal material, we have for the first time purified mRNA from naive, matured CNS axons, and identified the presence of >300 mRNA transcripts. We demonstrate that the transcripts are axonal in nature, and that many of the transcripts present in uninjured CNS axons overlap with those previously identified in PNS injury-conditioned DRG axons. The axonal transcripts detected in matured cortical axons are enriched for protein translational machinery, transport, cytoskeletal components, and mitochondrial maintenance. We next investigated how the axonal mRNA pool changes after axotomy, revealing that numerous gene transcripts related to intracellular transport, mitochondria and the cytoskeleton show decreased localization 2 d after injury. In contrast, gene transcripts related to axonal targeting and synaptic function show increased localization in regenerating cortical axons, suggesting that there is an increased capacity for axonal outgrowth and targeting, and increased support for synapse formation and presynaptic function in regenerating CNS axons after injury. Our data demonstrate that CNS axons contain many mRNA species of diverse functions, and suggest that, like invertebrate and PNS axons, CNS axons synthesize proteins locally, maintaining a degree of autonomy from the cell body.


Asunto(s)
Axones/fisiología , Corteza Cerebral/fisiología , Regeneración Nerviosa/fisiología , ARN Mensajero/aislamiento & purificación , Animales , Axones/química , Axotomía , Células Cultivadas , Corteza Cerebral/química , Técnicas In Vitro , Neurogénesis/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley
11.
J Neurochem ; 115(1): 11-22, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20193041

RESUMEN

The adult subventricular zone (SVZ) is a potential source of precursor cells to replace neural cells lost during demyelination. To better understand the molecular events that regulate neural precursor cell responsiveness in this context we undertook a microarray and quantitative PCR based analysis of genes expressed within the SVZ during cuprizone-induced demyelination. We identified an up-regulation of the genes encoding bone morphogenic protein 4 (BMP4) and its receptors. Immunohistochemistry confirmed an increase in BMP4 protein levels and also showed an increase in phosphorylated SMAD 1/5/8, a key component of BMP4 signalling, during demyelination. In vitro analysis revealed that neural precursor cells isolated from demyelinated animals, as well as those treated with BMP4, produce more astrocytes. Similarly, there were increased numbers of astrocytes in vivo within the SVZ during demyelination. Intraventricular infusion of Noggin, an endogenous antagonist of BMP4, during cuprizone-induced demyelination reduced pSMAD1/5/8, decreased astrocyte numbers and increased oligodendrocyte numbers in the SVZ. Our results suggest that lineage commitment of SVZ neural precursor cells is altered during demyelination and that BMP signalling plays a role in this process.


Asunto(s)
Astrocitos/efectos de los fármacos , Proteínas Morfogenéticas Óseas/fisiología , Ventrículos Cerebrales/patología , Enfermedades Desmielinizantes/patología , Oligodendroglía/efectos de los fármacos , Transducción de Señal/fisiología , Animales , Antimetabolitos , Proteína Morfogenética Ósea 4/antagonistas & inhibidores , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/fisiología , Receptores de Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Morfogenéticas Óseas/genética , Encéfalo/citología , Encéfalo/inmunología , Bromodesoxiuridina , Proteínas Portadoras/farmacología , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Ventrículos Cerebrales/efectos de los fármacos , Cuprizona , Enfermedades Desmielinizantes/inducido químicamente , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Microdisección , Inhibidores de la Monoaminooxidasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
12.
Nat Biotechnol ; 25(8): 894-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17687370

RESUMEN

A wealth of molecular interaction data is available in the literature, ranging from large-scale datasets to a single interaction confirmed by several different techniques. These data are all too often reported either as free text or in tables of variable format, and are often missing key pieces of information essential for a full understanding of the experiment. Here we propose MIMIx, the minimum information required for reporting a molecular interaction experiment. Adherence to these reporting guidelines will result in publications of increased clarity and usefulness to the scientific community and will support the rapid, systematic capture of molecular interaction data in public databases, thereby improving access to valuable interaction data.


Asunto(s)
Bases de Datos de Proteínas/normas , Guías como Asunto , Almacenamiento y Recuperación de la Información/normas , Mapeo de Interacción de Proteínas/normas , Proteómica/normas , Investigación/normas , Humanos , Internacionalidad
13.
Biol Psychiatry ; 88(12): 898-909, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32331824

RESUMEN

BACKGROUND: Alcohol use disorder (AUD) is a major socioeconomic burden on society, and current pharmacotherapeutic treatment options are inadequate. Aberrant alcohol use and seeking alters frontostriatal function. METHODS: We performed genome-wide RNA sequencing and subsequent quantitative polymerase chain reaction and receptor binding validation in the caudate-putamen of human AUD samples to identify potential therapeutic targets. We then back-translated our top candidate targets into a rodent model of long-term alcohol consumption to assess concordance of molecular adaptations in the rat striatum. Finally, we adopted rat behavioral models of alcohol intake and seeking to validate a potential therapeutic target. RESULTS: We found that G protein-coupled receptors were the top canonical pathway differentially regulated in individuals with AUD. The M4 muscarinic acetylcholine receptor (mAChR) was downregulated at the gene and protein levels in the putamen, but not in the caudate, of AUD samples. We found concordant downregulation of the M4 mAChR, specifically on dopamine D1 receptor-expressing medium spiny neurons in the rat dorsolateral striatum. Systemic administration of the selective M4 mAChR positive allosteric modulator, VU0467154, reduced home cage and operant alcohol self-administration, motivation to obtain alcohol, and cue-induced reinstatement of alcohol seeking in rats. Local microinjections of VU0467154 in the rat dorsolateral striatum reduced alcohol self-administration and cue-induced reinstatement of alcohol seeking. CONCLUSIONS: Collectively, these results identify the M4 mAChR as a potential therapeutic target for the treatment of AUD and the D1 receptor-positive medium spiny neurons in the dorsolateral striatum as a key site mediating the actions of M4 mAChR in relation to alcohol consumption and seeking.


Asunto(s)
Alcoholismo , Receptor Muscarínico M4 , Acetilcolina , Alcoholismo/tratamiento farmacológico , Alcoholismo/genética , Animales , Colinérgicos , Humanos , Ratas , Receptor Muscarínico M4/genética , Roedores
14.
Neurosignals ; 17(4): 311-27, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19816067

RESUMEN

The central nervous system (CNS) displays heterogeneity at regional, cellular and subcellular levels, making analysis of transcriptomic events accompanying neural injury particularly challenging. Microarray technology provides methods for elucidating global changes in neural gene expression and discovery of signalling pathways within this complex biological network. The lack of suitable and sufficient human CNS tissue along with its inherent variability means that diverse animal models of both multiple sclerosis and neurotrauma are vital for examining the pathophysiological changes accompanying neural injury resulting from disease or trauma. Gene expression profiling of these models is providing valuable information about mechanisms of damage, repair and regeneration and candidate treatments. In vitro models of neural injury are also proving useful, and transcriptomics is enhancing our understanding of the properties of neural stem cells with a view to their therapeutic application in neural repair. Thoughtful experimental design and analysis of microarray experiments is crucial for extracting biological meaning from the vast amount of data produced. In this review we discuss the current and emerging application of transcriptomics for the study of neural function in health, disease and injury.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/lesiones , Sistema Nervioso Central/metabolismo , Perfilación de la Expresión Génica/métodos , Perfilación de la Expresión Génica/tendencias , Animales , Investigación Biomédica/métodos , Investigación Biomédica/tendencias , Humanos , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Regeneración Nerviosa/fisiología , Neuronas/metabolismo , Células Madre/metabolismo
15.
Proteomics ; 8(9): 1819-31, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18384105

RESUMEN

Neurodegenerative illnesses are characterized by aberrant metabolism of biometals such as copper (Cu), zinc (Zn) and iron (Fe). However, little is known about the metabolic effects associated with altered metal homeostasis. In this study, we used an in vitro model of altered Cu homeostasis to investigate how Cu regulates cellular protein expression. Human fibroblasts containing a natural deletion mutation of the Menkes (MNK) ATP7A Cu transporter (MNK deleted) were compared to fibroblasts overexpressing ATP7A (MNK transfected). Cultures of MNK-transfected (Low-Cu) cells exhibited 95% less intracellular Cu than MNK-deleted (High-Cu) cells. Comparative proteomic analysis of the two cell-lines was performed using antibody microarrays, and significant differential protein expression was observed between Low-Cu and High-Cu cell-lines. Western blot analysis confirmed the altered protein expression of Ku80, nexilin, L-caldesmon, MAP4, Inhibitor 2 and DNA topoisomerase I. The top 50 altered proteins were analysed using the software program Pathway Studio (Ariadne Genomics) and revealed a significant over-representation of proteins involved in DNA repair and maintenance. Further analysis confirmed that expression of the DNA repair protein Ku80 was dependent on cellular Cu homeostasis and that Low-Cu levels in fibroblasts resulted in elevated susceptibility to DNA oxidation.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Transporte de Catión/genética , Cobre/química , Fibroblastos/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Antígenos Nucleares/biosíntesis , Transporte Biológico , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/metabolismo , Biología Computacional/métodos , ATPasas Transportadoras de Cobre , ADN/química , Proteínas de Unión al ADN/biosíntesis , Humanos , Autoantígeno Ku , Enfermedades Neurodegenerativas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxígeno/química , Análisis por Matrices de Proteínas , Proteómica/métodos , Programas Informáticos
16.
Front Mol Neurosci ; 11: 433, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30618600

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by a tandem repeat mutation encoding an expanded polyglutamine tract in the huntingtin protein, which leads to cognitive, psychiatric and motor dysfunction. Exposure to environmental enrichment (EE), which enhances levels of cognitive stimulation and physical activity, has therapeutic effects on cognitive, affective and motor function of transgenic HD mice. The present study investigated gene expression changes and behavioral pharmacology in male and female R6/1 transgenic HD mice at an early time-point in HD progression associated with onset of cognitive and affective abnormalities, following EE and exercise (wheel running) interventions. We have demonstrated changes in expression levels of the serotonin (5-HT) receptor Htr1a, Htr1b, Htr2a and Htr2c genes (encoding the 5-HT1A, 5-HT1B, 5-HT2A and 5-HT2C receptors, respectively) in HD brains at 8 weeks of age, using quantitative real-time PCR. In contrast, expression of the serotonin transporter (SerT, also known as 5-HTT or Slc6a4) was not altered in these brains. Furthermore, we identified region-specific, sex-specific and environmentally regulated (comparing EE, exercise and standard housing conditions) impacts on gene expression of particular 5-HT receptors, as well as SerT. For example, SerT gene expression was upregulated by exercise (wheel running from 6 to 8 weeks of age) in the hippocampus. Interestingly, when EE was introduced from 6 to 8 weeks of age, Htr2a gene expression was upregulated in the cortex, striatum and hippocampus of male mice. EE also rescued the functional activity of 5-HT2 receptors as observed in the head-twitch test, reflecting sexually dimorphic effects of environmental stimulation. These findings demonstrate that disruption of the serotonergic system occurs early in HD pathogenesis and, together with previous findings, show that the timing and duration of environmental interventions are critical in terms of their ability to modify gene expression. This study is the first to show that EE is able to selectively enhance both gene expression of a neurotransmitter receptor and the functional consequences on behavioral pharmacology, and links this molecular modulation to the therapeutic effects of environmental stimulation in this neurodegenerative disease.

17.
Neurochem Int ; 50(2): 336-44, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17118492

RESUMEN

The expression levels of a number of genes associated with inflammation and immune function change with advancing age. Melatonin modulates gene expression levels of several of these genes. Therefore the declining levels of melatonin associated with age may play a role in the physiological effects of aging. We used oligonucleotide microarrays to measure age-related changes in mRNA expression in the murine CNS, and to study the effect of prolonged administration of dietary melatonin upon these changes. CB6F1 male mice were fed 40 ppm melatonin for 2.1 months prior to sacrifice at age 26.5 months, and compared with both age-matched controls and young, 4.5-month-old untreated controls. Total RNA was extracted from whole brain (excluding cerebellum and brain stem) and individual samples were hybridized to Affymetrix Mouse 430-2.0 arrays. The expression of a substantial number of genes was modulated by melatonin treatment and changes in selected genes were validated by quantitative reverse transcription polymerase chain reaction (qRT-PCR). A subset of these genes did not change with age. Conversely, some genes modulated by age were also modulated by melatonin treatment. In general, melatonin treatment drove the expression levels of these genes closer to the expression levels detected in the younger animals. Notably, the abundance of lipocalin 2 (Lcn2) mRNA increased with age and was decreased in old animals treated with melatonin. Lcn2 is a member of the acute phase response family of proteins and its mRNA levels in the brain increase in response to inflammation. Many of the genes with expression reduced by melatonin are involved in inflammation and the immune system. This suggests that melatonin treatment may influence the inflammatory responses of old animals, driving them to resemble more closely those occurring in young animals.


Asunto(s)
Envejecimiento/fisiología , Sistema Nervioso Central/metabolismo , Expresión Génica/efectos de los fármacos , Melatonina/farmacología , Proteínas de Fase Aguda/biosíntesis , Animales , N-Acetiltransferasa de Arilalquilamina/biosíntesis , Sistema Nervioso Central/efectos de los fármacos , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Lipocalina 2 , Lipocalinas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor de Melatonina MT1/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Neurosci ; 25(17): 4217-21, 2005 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-15858047

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which there are few therapeutics that affect the underlying disease mechanism. Recent epidemiological studies, however, suggest that lifestyle changes may slow the onset/progression of AD. Here we have used TgCRND8 mice to examine directly the interaction between exercise and the AD cascade. Five months of voluntary exercise resulted in a decrease in extracellular amyloid-beta (Abeta) plaques in the frontal cortex (38%; p = 0.018), the cortex at the level of the hippocampus (53%; p = 0.0003), and the hippocampus (40%; p = 0.06). This was associated with decreased cortical Abeta1-40 (35%; p = 0.005) and Abeta1-42 (22%; p = 0.04) (ELISA). The mechanism appears to be mediated by a change in the processing of the amyloid precursor protein (APP) after short-term exercise, because 1 month of activity decreased the proteolytic fragments of APP [for alpha-C-terminal fragment (alpha-CTF), 54% and p = 0.04; for beta-CTF, 35% and p = 0.03]. This effect was independent of mRNA/protein changes in neprilysin and insulin-degrading enzyme and, instead, may involve neuronal metabolism changes that are known to affect APP processing and to be regulated by exercise. Long-term exercise also enhanced the rate of learning of TgCRND8 animals in the Morris water maze, with significant (p < 0.02) reductions in escape latencies over the first 3 (of 6) trial days. In support of existing epidemiological studies, this investigation demonstrates that exercise is a simple behavioral intervention sufficient to inhibit the normal progression of AD-like neuropathology in the TgCRND8 mouse model.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/rehabilitación , Amiloide/metabolismo , Condicionamiento Físico Animal/fisiología , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Ácido Aspártico Endopeptidasas , Western Blotting/métodos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Endopeptidasas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Inmunohistoquímica/métodos , Masculino , Ratones , Ratones Transgénicos , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo
19.
J Alzheimers Dis ; 51(2): 471-87, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26890748

RESUMEN

BACKGROUND: There is strong evidence that iron homeostasis is impaired in the aging and Alzheimer's disease (AD) brain and that this contributes to neurodegeneration. Apolipoprotein E (APOE) has been identified as the strongest genetic risk factor for AD. However, the interaction between the two has yet to be fully explored. OBJECTIVE: This study aimed to investigate the relationship between exogenous iron levels and ApoE in neurons and astrocytes. METHODS: Our study used primary cultured cortical neurons and astrocytes to investigate the changes in ApoE caused by iron. Western blot and RT-PCR were used to measure ApoE. RESULTS: We observed that iron upregulated intracellular ApoE levels in both neurons and astrocytes at the post-transcriptional and transcriptional level, respectively. However, there was less full-length ApoE secreted by neurons and astrocytes after iron treatment. We speculate that this might impair brain lipid metabolism and amyloid-ß clearance. In terms of ApoE receptors, we observed that neuronal LRP-1 levels were increased by the addition of exogenous iron, which could contribute to AßPP endocytosis in neurons. However, there were no significant changes in neuronal LDLR, astrocyte LDLR, or astrocyte LRP-1. CONCLUSION: Our study reveals that iron may contribute to the pathogenesis of AD by affecting ApoE and its receptors and supports the notion that iron chelation should be investigated as a therapeutic strategy for AD.


Asunto(s)
Apolipoproteínas E/metabolismo , Astrocitos/metabolismo , Hierro/metabolismo , Neuronas/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Cobre/metabolismo , Ferritinas/metabolismo , Inmunohistoquímica , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Zinc/metabolismo
20.
Neurobiol Aging ; 26(4): 511-20, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15653179

RESUMEN

Voluntary exercise increases hippocampal brain-derived neurotrophic factor (BDNF) expression in young animals. In this investigation we examined the induction of BDNF protein in the hippocampus of young (2 months), late middle-aged (15 months) and old (24 months) animals over 4 weeks of exercise. Average running distances decreased with age, with the old animals also maintaining a constant level of activity over time, whereas the other groups tended to increase their average running distance. All animals demonstrated a biphasic profile of BDNF protein induction, with a significant (P<0.05) increase after 1 week of exercise followed by a decrease to near sedentary levels at 2 weeks. After this, BDNF protein levels increased significantly (P<0.05), as compared to baseline, primarily only in the young animals. In whole hippocampal homogenates, only particular BDNF mRNA exons were significantly (P<0.05) changed as a result of exercise, with the largest induction occurring in young animals. BDNF protein induction may, therefore, not be directly correlated with significant mRNA changes. Exercise may represent a therapeutic tool for disorders which involve a decrease in BDNF.


Asunto(s)
Envejecimiento/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Regulación de la Expresión Génica/fisiología , Hipocampo/metabolismo , Condicionamiento Físico Animal/fisiología , Factores de Edad , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Ensayo de Inmunoadsorción Enzimática/métodos , Exones , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA