Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Acta Neuropathol ; 146(3): 451-475, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37488208

RESUMEN

Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease mainly affecting upper and lower motoneurons. Several functionally heterogeneous genes have been associated with the familial form of this disorder (fALS), depicting an extremely complex pathogenic landscape. This heterogeneity has limited the identification of an effective therapy, and this bleak prognosis will only improve with a greater understanding of convergent disease mechanisms. Recent evidence from human post-mortem material and diverse model systems has highlighted the synapse as a crucial structure actively involved in disease progression, suggesting that synaptic aberrations might represent a shared pathological feature across the ALS spectrum. To test this hypothesis, we performed the first comprehensive analysis of the synaptic proteome from post-mortem spinal cord and human iPSC-derived motoneurons carrying mutations in the major ALS genes. This integrated approach highlighted perturbations in the molecular machinery controlling vesicle release as a shared pathomechanism in ALS. Mechanistically, phosphoproteomic analysis linked the presynaptic vesicular phenotype to an accumulation of cytotoxic protein aggregates and to the pro-apoptotic activation of the transcription factor c-Jun, providing detailed insights into the shared pathobiochemistry in ALS. Notably, sub-chronic treatment of our iPSC-derived motoneurons with the fatty acid docosahexaenoic acid exerted a neuroprotective effect by efficiently rescuing the alterations revealed by our multidisciplinary approach. Together, this study provides strong evidence for the central and convergent role played by the synaptic microenvironment within the ALS spinal cord and highlights a potential therapeutic target that counteracts degeneration in a heterogeneous cohort of human motoneuron cultures.


Asunto(s)
Esclerosis Amiotrófica Lateral , Enfermedades Neurodegenerativas , Humanos , Esclerosis Amiotrófica Lateral/patología , Enfermedades Neurodegenerativas/patología , Proteómica , Superóxido Dismutasa-1/genética , Neuronas Motoras/metabolismo
2.
Alzheimers Dement ; 19(6): 2560-2574, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36547260

RESUMEN

INTRODUCTION: It remains unclear why age increases risk of Alzheimer's disease and why some people experience age-related cognitive decline in the absence of dementia. Here we test the hypothesis that resilience to molecular changes in synapses contribute to healthy cognitive ageing. METHODS: We examined post-mortem brain tissue from people in mid-life (n = 15), healthy ageing with either maintained cognition (n = 9) or lifetime cognitive decline (n = 8), and Alzheimer's disease (n = 13). Synapses were examined with high resolution imaging, proteomics, and RNA sequencing. Stem cell-derived neurons were challenged with Alzheimer's brain homogenate. RESULTS: Synaptic pathology increased, and expression of genes involved in synaptic signaling decreased between mid-life, healthy ageing and Alzheimer's. In contrast, brain tissue and neurons from people with maintained cognition during ageing exhibited decreases in synaptic signaling genes compared to people with cognitive decline. DISCUSSION: Efficient synaptic networks without pathological protein accumulation may contribute to maintained cognition during ageing.


Asunto(s)
Enfermedad de Alzheimer , Envejecimiento Cognitivo , Envejecimiento Saludable , Sinapsis , Cognición , Sinapsis/metabolismo , Sinapsis/patología , Encéfalo/metabolismo , Encéfalo/patología , Análisis de Secuencia de ARN , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuronas/metabolismo , Neuronas/patología , Transmisión Sináptica , Cambios Post Mortem , Envejecimiento Saludable/metabolismo , Envejecimiento Saludable/patología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Humanos , Masculino , Femenino , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Gliosis/patología
3.
Hum Mol Genet ; 29(16): 2674-2683, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32644120

RESUMEN

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by mutations in survival motor neuron 1 (SMN1). SMN-restoring therapies have recently emerged; however, preclinical and clinical studies revealed a limited therapeutic time window and systemic aspects of the disease. This raises a fundamental question of whether SMA has presymptomatic, developmental components to disease pathogenesis. We have addressed this by combining micro-computed tomography (µCT) and comparative proteomics to examine systemic pre-symptomatic changes in a prenatal mouse model of SMA. Quantitative µCT analyses revealed that SMA embryos were significantly smaller than littermate controls, indicative of general developmental delay. More specifically, cardiac ventricles were smaller in SMA hearts, whilst liver and brain remained unaffected. In order to explore the molecular consequences of SMN depletion during development, we generated comprehensive, high-resolution, proteomic profiles of neuronal and non-neuronal organs in SMA mouse embryos. Significant molecular perturbations were observed in all organs examined, highlighting tissue-specific prenatal molecular phenotypes in SMA. Together, our data demonstrate considerable systemic changes at an early, presymptomatic stage in SMA mice, revealing a significant developmental component to SMA pathogenesis.


Asunto(s)
Atrofia Muscular Espinal/genética , Miocardio/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Corazón/fisiopatología , Humanos , Hígado/metabolismo , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/diagnóstico , Atrofia Muscular Espinal/patología , Miocardio/patología , Fenotipo , Diagnóstico Prenatal , Proteómica , Microtomografía por Rayos X
4.
PLoS Pathog ; 15(2): e1007561, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30742688

RESUMEN

Paramyxoviruses can establish persistent infections both in vitro and in vivo, some of which lead to chronic disease. However, little is known about the molecular events that contribute to the establishment of persistent infections by RNA viruses. Using parainfluenza virus type 5 (PIV5) as a model we show that phosphorylation of the P protein, which is a key component of the viral RNA polymerase complex, determines whether or not viral transcription and replication becomes repressed at late times after infection. If the virus becomes repressed, persistence is established, but if not, the infected cells die. We found that single amino acid changes at various positions within the P protein switched the infection phenotype from lytic to persistent. Lytic variants replicated to higher titres in mice than persistent variants and caused greater infiltration of immune cells into infected lungs but were cleared more rapidly. We propose that during the acute phases of viral infection in vivo, lytic variants of PIV5 will be selected but, as the adaptive immune response develops, variants in which viral replication can be repressed will be selected, leading to the establishment of prolonged, persistent infections. We suggest that similar selection processes may operate for other RNA viruses.


Asunto(s)
Infecciones por Paramyxoviridae/genética , Paramyxoviridae/genética , Fosfoproteínas/genética , Proteínas Virales/genética , Células A549 , Sustitución de Aminoácidos/genética , Animales , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/patogenicidad , Paramyxoviridae/patogenicidad , Infecciones por Paramyxoviridae/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiología , Fosforilación , ARN Viral , Proteínas Virales/metabolismo , Proteínas Virales/fisiología , Replicación Viral
5.
Int J Mol Sci ; 22(22)2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34830029

RESUMEN

Cyst nematodes are important herbivorous pests in agriculture that obtain nutrients through specialized root structures termed syncytia. Syncytium initiation, development, and functioning are a research focus because syncytia are the primary interface for molecular interactions between the host plant and parasite. The small size and complex development (over approximately two weeks) of syncytia hinder precise analyses, therefore most studies have analyzed the transcriptome of infested whole-root systems or syncytia-containing root segments. Here, we describe an effective procedure to microdissect syncytia induced by Globodera rostochiensis from tomato roots and to analyze the syncytial proteome using mass spectrometry. As little as 15 mm2 of 10-µm-thick sections dissected from 30 syncytia enabled the identification of 100-200 proteins in each sample, indicating that mass-spectrometric methods currently in use achieved acceptable sensitivity for proteome profiling of microscopic samples of plant tissues (approximately 100 µg). Among the identified proteins, 48 were specifically detected in syncytia and 7 in uninfected roots. The occurrence of approximately 50% of these proteins in syncytia was not correlated with transcript abundance estimated by quantitative reverse-transcription PCR analysis. The functional categories of these proteins confirmed that protein turnover, stress responses, and intracellular trafficking are important components of the proteome dynamics of developing syncytia.


Asunto(s)
Chromadorea , Células Gigantes/metabolismo , Proteínas de Plantas/metabolismo , Raíces de Plantas , Proteoma/metabolismo , Solanum lycopersicum , Animales , Solanum lycopersicum/metabolismo , Solanum lycopersicum/parasitología , Raíces de Plantas/metabolismo , Raíces de Plantas/parasitología
6.
Neurobiol Dis ; 130: 104496, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31176719

RESUMEN

Neurodegenerative and neuromuscular disorders can manifest throughout the lifespan of an individual, from infant to elderly individuals. Axonal and synaptic degeneration are early and critical elements of nearly all human neurodegenerative diseases and neural injury, however the molecular mechanisms which regulate this process are yet to be fully elucidated. Furthermore, how the molecular mechanisms governing degeneration are impacted by the age of the individual is poorly understood. Interestingly, in mice which are under 3 weeks of age, the degeneration of axons and synapses following hypoxic or traumatic injury is significantly slower. This process, known as Wallerian degeneration (WD), is a molecularly and morphologically distinct subtype of neurodegeneration by which axons and synapses undergo distinct fragmentation and death following a range of stimuli. In this study, we first use an ex-vivo model of axon injury to confirm the significant delay in WD in neonatal mice. We apply tandem mass-tagging quantitative proteomics to profile both nerve and muscle between P12 and P24 inclusive. Application of unbiased in silico workflows to relevant protein identifications highlights a steady elevation in oxidative phosphorylation cascades corresponding to the accelerated degeneration rate. We demonstrate that inhibition of Complex I prevents the axotomy-induced rise in reactive oxygen species and protects axons following injury. Furthermore, we reveal that pharmacological activation of oxidative phosphorylation significantly accelerates degeneration at the neuromuscular junction in neonatal mice. In summary, we reveal dramatic changes in the neuromuscular proteome during post-natal maturation of the neuromuscular system, and demonstrate that endogenous dynamics in mitochondrial bioenergetics during this time window have a functional impact upon regulating the stability of the neuromuscular system.


Asunto(s)
Mitocondrias/metabolismo , Unión Neuromuscular/metabolismo , Fosforilación Oxidativa , Degeneración Walleriana/metabolismo , Animales , Animales Recién Nacidos , Ratones , Ratones Endogámicos C57BL , Unión Neuromuscular/patología , Degeneración Walleriana/patología
7.
J Cell Sci ; 130(2): 325-331, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28049716

RESUMEN

α-Synuclein plays a central role in Parkinson's disease, where it contributes to the vulnerability of synapses to degeneration. However, the downstream mechanisms through which α-synuclein controls synaptic stability and degeneration are not fully understood. Here, comparative proteomics on synapses isolated from α-synuclein-/- mouse brain identified mitochondrial proteins as primary targets of α-synuclein, revealing 37 mitochondrial proteins not previously linked to α-synuclein or neurodegeneration pathways. Of these, sideroflexin 3 (SFXN3) was found to be a mitochondrial protein localized to the inner mitochondrial membrane. Loss of SFXN3 did not disturb mitochondrial electron transport chain function in mouse synapses, suggesting that its function in mitochondria is likely to be independent of canonical bioenergetic pathways. In contrast, experimental manipulation of SFXN3 levels disrupted synaptic morphology at the Drosophila neuromuscular junction. These results provide novel insights into α-synuclein-dependent pathways, highlighting an important influence on mitochondrial proteins at the synapse, including SFXN3. We also identify SFXN3 as a new mitochondrial protein capable of regulating synaptic morphology in vivo.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Sinapsis/metabolismo , alfa-Sinucleína/metabolismo , Animales , Drosophila melanogaster/metabolismo , Metabolismo Energético , Ontología de Genes , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Membranas Mitocondriales/metabolismo , Unión Neuromuscular/metabolismo
8.
Nat Chem Biol ; 13(5): 514-521, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28288108

RESUMEN

Inducing macromolecular interactions with small molecules to activate cellular signaling is a challenging goal. PROTACs (proteolysis-targeting chimeras) are bifunctional molecules that recruit a target protein in proximity to an E3 ubiquitin ligase to trigger protein degradation. Structural elucidation of the key ternary ligase-PROTAC-target species and its impact on target degradation selectivity remain elusive. We solved the crystal structure of Brd4 degrader MZ1 in complex with human VHL and the Brd4 bromodomain (Brd4BD2). The ligand folds into itself to allow formation of specific intermolecular interactions in the ternary complex. Isothermal titration calorimetry studies, supported by surface mutagenesis and proximity assays, are consistent with pronounced cooperative formation of ternary complexes with Brd4BD2. Structure-based-designed compound AT1 exhibits highly selective depletion of Brd4 in cells. Our results elucidate how PROTAC-induced de novo contacts dictate preferential recruitment of a target protein into a stable and cooperative complex with an E3 ligase for selective degradation.


Asunto(s)
Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteolisis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Proteínas de Ciclo Celular , Cristalografía por Rayos X , Dipéptidos/química , Dipéptidos/farmacología , Elonguina , Compuestos Heterocíclicos con 3 Anillos/química , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Termodinámica , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/química , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
9.
Mol Cell Biochem ; 456(1-2): 157-165, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30694515

RESUMEN

Stroke is a common disorder with significant morbidity and mortality, and complex aetiology involving both environmental and genetic risk factors. Although some of the major risk factors for stoke, such as smoking and hypertension, are well-documented, the underlying genetic and detailed molecular mechanisms remain elusive. Exploring the relevant biochemical pathways may contribute to the clinical diagnosis of stroke and shed light on its aetiology. A comparative proteomic analysis of blood serum of a pair of monozygotic (MZ) twins discordant for ischaemic stroke (IS) was performed using a label-free quantitative proteomics approach. To overcome the limit of reproducibility in the serum preparation, two separate runs were performed, each consisting of three technical replicates per sample. Biological processes associated with proteins differentially expressed between the twins were explored with gene ontology (GO) classification using the functional analysis tool g:Profiler. ANOVA test performed in Progenesis LC-MS identified 179 (run 1) and 209 (run 2) proteins as differentially expressed between the affected and unaffected twin (p < 0.05). Furthermore, the level of serum fibulin 1, an extracellular matrix protein associated with arterial stiffness, was on average 13.37-fold higher in the affected twin. Each dataset was then analysed independently, and the proteins were classified according to GO terms. The categories overrepresented in the affected twin predominantly corresponded to stroke-relevant processes, including wound healing, blood coagulation and haemostasis, with a high proportion of the proteins overexpressed in the affected twin associated with these terms. By contrast, in the unaffected twin diagnosed with atopic dermatitis, there were increased levels of keratin proteins and GO terms associated with skin development. The identification of cellular pathways enriched in IS as well as the upregulation of fibulin 1 sheds new light on the underlying disease-causing mechanisms at the molecular level. Our findings of distinct proteomic signatures associated with IS and atopic dermatitis suggest proteomic profiling could be used as a general approach for improved diagnostic, prognostic and therapeutic strategies.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Isquemia Encefálica/metabolismo , Proteoma/metabolismo , Proteómica , Accidente Cerebrovascular/metabolismo , Gemelos Monocigóticos , Femenino , Humanos , Masculino
10.
Brain ; 141(10): 2878-2894, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30239612

RESUMEN

Deafferentation of motor neurons as a result of defective sensory-motor connectivity is a critical early event in the pathogenesis of spinal muscular atrophy, but the underlying molecular pathways remain unknown. We show that restoration of ubiquitin-like modifier-activating enzyme 1 (UBA1) was sufficient to correct sensory-motor connectivity in the spinal cord of mice with spinal muscular atrophy. Aminoacyl-tRNA synthetases, including GARS, were identified as downstream targets of UBA1. Regulation of GARS by UBA1 occurred via a non-canonical pathway independent of ubiquitylation. Dysregulation of UBA1/GARS pathways in spinal muscular atrophy mice disrupted sensory neuron fate, phenocopying GARS-dependent defects associated with Charcot-Marie-Tooth disease. Sensory neuron fate was corrected following restoration of UBA1 expression and UBA1/GARS pathways in spinal muscular atrophy mice. We conclude that defective sensory motor connectivity in spinal muscular atrophy results from perturbations in a UBA1/GARS pathway that modulates sensory neuron fate, thereby highlighting significant molecular and phenotypic overlap between spinal muscular atrophy and Charcot-Marie-Tooth disease.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Atrofia Muscular Espinal/patología , Vías Nerviosas/patología , Enzimas Activadoras de Ubiquitina/metabolismo , Animales , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Regulación de la Expresión Génica/fisiología , Células HEK293 , Humanos , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/metabolismo , Vías Nerviosas/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/patología , Transducción de Señal/fisiología , Médula Espinal/metabolismo , Médula Espinal/patología
12.
Mol Cell Proteomics ; 14(3): 596-608, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25561500

RESUMEN

The lipid raft concept proposes that membrane environments enriched in cholesterol and sphingolipids cluster certain proteins and form platforms to integrate cell signaling. In cardiac muscle, caveolae concentrate signaling molecules and ion transporters, and play a vital role in adrenergic regulation of excitation-contraction coupling, and consequently cardiac contractility. Proteomic analysis of cardiac caveolae is hampered by the presence of contaminants that have sometimes, erroneously, been proposed to be resident in these domains. Here we present the first unbiased analysis of the proteome of cardiac caveolae, and investigate dynamic changes in their protein constituents following adrenoreceptor (AR) stimulation. Rat ventricular myocytes were treated with methyl-ß-cyclodextrin (MßCD) to deplete cholesterol and disrupt caveolae. Buoyant caveolin-enriched microdomains (BCEMs) were prepared from MßCD-treated and control cell lysates using a standard discontinuous sucrose gradient. BCEMs were harvested, pelleted, and resolubilized, then alkylated, digested, and labeled with iTRAQ reagents, and proteins identified by LC-MS/MS on a LTQ Orbitrap Velos Pro. Proteins were defined as BCEM resident if they were consistently depleted from the BCEM fraction following MßCD treatment. Selective activation of α-, ß1-, and ß2-AR prior to preparation of BCEMs was achieved by application of agonist/antagonist pairs for 10 min in populations of field-stimulated myocytes. We typically identified 600-850 proteins per experiment, of which, 249 were defined as high-confidence BCEM residents. Functional annotation clustering indicates cardiac BCEMs are enriched in integrin signaling, guanine nucleotide binding, ion transport, and insulin signaling clusters. Proteins possessing a caveolin binding motif were poorly enriched in BCEMs, suggesting this is not the only mechanism that targets proteins to caveolae. With the notable exception of the cavin family, very few proteins show altered abundance in BCEMs following AR activation, suggesting signaling complexes are preformed in BCEMs to ensure a rapid and high fidelity response to adrenergic stimulation in cardiac muscle.


Asunto(s)
Agonistas Adrenérgicos/farmacología , Caveolas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Proteoma/aislamiento & purificación , Proteómica/métodos , Antagonistas Adrenérgicos/farmacología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Ratas , Transducción de Señal , beta-Ciclodextrinas/farmacología
13.
Mol Cell Proteomics ; 14(11): 3072-86, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26364976

RESUMEN

Equine grass sickness (EGS) is an acute, predominantly fatal, multiple system neuropathy of grazing horses with reported incidence rates of ∼2%. An apparently identical disease occurs in multiple species, including but not limited to cats, dogs, and rabbits. Although the precise etiology remains unclear, ultrastructural findings have suggested that the primary lesion lies in the glycoprotein biosynthetic pathway of specific neuronal populations. The goal of this study was therefore to identify the molecular processes underpinning neurodegeneration in EGS. Here, we use a bottom-up approach beginning with the application of modern proteomic tools to the analysis of cranial (superior) cervical ganglion (CCG, a consistently affected tissue) from EGS-affected patients and appropriate control cases postmortem. In what appears to be the proteomic application of modern proteomic tools to equine neuronal tissues and/or to an inherent neurodegenerative disease of large animals (not a model of human disease), we identified 2,311 proteins in CCG extracts, with 320 proteins increased and 186 decreased by greater than 20% relative to controls. Further examination of selected proteomic candidates by quantitative fluorescent Western blotting (QFWB) and subcellular expression profiling by immunohistochemistry highlighted a previously unreported dysregulation in proteins commonly associated with protein misfolding/aggregation responses seen in a myriad of human neurodegenerative conditions, including but not limited to amyloid precursor protein (APP), microtubule associated protein (Tau), and multiple components of the ubiquitin proteasome system (UPS). Differentially expressed proteins eligible for in silico pathway analysis clustered predominantly into the following biofunctions: (1) diseases and disorders, including; neurological disease and skeletal and muscular disorders and (2) molecular and cellular functions, including cellular assembly and organization, cell-to-cell signaling and interaction (including epinephrine, dopamine, and adrenergic signaling and receptor function), and small molecule biochemistry. Interestingly, while the biofunctions identified in this study may represent pathways underpinning EGS-induced neurodegeneration, this is also the first demonstration of potential molecular conservation (including previously unreported dysregulation of the UPS and APP) spanning the degenerative cascades from an apparently unrelated condition of large animals, to small animal models with altered neuronal vulnerability, and human neurological conditions. Importantly, this study highlights the feasibility and benefits of applying modern proteomic techniques to veterinary investigations of neurodegenerative processes in diseases of large animals.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Enfermedades de los Caballos/genética , Enfermedades Neurodegenerativas/genética , Deficiencias en la Proteostasis/genética , Ubiquitina/genética , Proteínas tau/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Femenino , Ganglios Sensoriales/química , Ganglios Sensoriales/metabolismo , Ganglios Sensoriales/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Enfermedades de los Caballos/diagnóstico , Enfermedades de los Caballos/metabolismo , Enfermedades de los Caballos/patología , Caballos , Masculino , Anotación de Secuencia Molecular , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica , Deficiencias en la Proteostasis/diagnóstico , Deficiencias en la Proteostasis/metabolismo , Deficiencias en la Proteostasis/patología , Ubiquitina/metabolismo , Proteínas tau/metabolismo
14.
Proc Natl Acad Sci U S A ; 111(49): 17432-7, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25326418

RESUMEN

The siRNA knockdown of IFN Regulatory Factor 5 (IRF5) in the human plasmacytoid dendritic cell line Gen2.2 prevented IFNß production induced by compound CL097, a ligand for Toll-like receptor 7 (TLR7). CL097 also stimulated the phosphorylation of IRF5 at Ser462 and stimulated the nuclear translocation of wild-type IRF5, but not the IRF5[Ser462Ala] mutant. The CL097-stimulated phosphorylation of IRF5 at Ser462 and its nuclear translocation was prevented by the pharmacological inhibition of protein kinase IKKß or the siRNA knockdown of IKKß or its "upstream" activator, the protein kinase TAK1. Similar results were obtained in a murine macrophage cell line stimulated with the TLR7 agonist compound R848 or the nucleotide oligomerization domain 1 (NOD1) agonist KF-1B. IKKß phosphorylated IRF5 at Ser462 in vitro and induced the dimerization of wild-type IRF5 but not the IRF5[S462A] mutant. These findings demonstrate that IKKß activates two "master" transcription factors of the innate immune system, IRF5 and NF-κB.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteínas I-kappa B/metabolismo , Factores Reguladores del Interferón/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Humanos , Inmunidad Innata , Inflamación , Interferón beta/metabolismo , Ligandos , Ratones , Microscopía Fluorescente , Mutación , Fosforilación , Multimerización de Proteína , Serina/química , Transcripción Genética , Transfección
15.
PLoS Genet ; 10(6): e1004390, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24901984

RESUMEN

Nuclear myosin 1c (NM1) mediates RNA polymerase I (pol I) transcription activation and cell cycle progression by facilitating PCAF-mediated H3K9 acetylation, but the molecular mechanism by which NM1 is regulated remains unclear. Here, we report that at early G1 the glycogen synthase kinase (GSK) 3ß phosphorylates and stabilizes NM1, allowing for NM1 association with the chromatin. Genomic analysis by ChIP-Seq showed that this mechanism occurs on the rDNA as active GSK3ß selectively occupies the gene. ChIP assays and transmission electron microscopy in GSK3ß-/- mouse embryonic fibroblasts indicated that at G1 rRNA synthesis is suppressed due to decreased H3K9 acetylation leading to a chromatin state incompatible with transcription. We found that GSK3ß directly phosphorylates the endogenous NM1 on a single serine residue (Ser-1020) located within the NM1 C-terminus. In G1 this phosphorylation event stabilizes NM1 and prevents NM1 polyubiquitination by the E3 ligase UBR5 and proteasome-mediated degradation. We conclude that GSK3ß-mediated phosphorylation of NM1 is required for pol I transcription activation.


Asunto(s)
Fase G1/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Miosina Tipo I/metabolismo , Activación Transcripcional/genética , Ubiquitina-Proteína Ligasas/metabolismo , Acetilación , Animales , Línea Celular , Cromatina/genética , ADN Ribosómico/genética , Proteínas F-Box/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Células HeLa , Histonas/metabolismo , Humanos , Ratones , Ratones Noqueados , Fosforilación , Proteolisis , Interferencia de ARN , ARN Polimerasa I/genética , ARN Interferente Pequeño , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Factores de Transcripción p300-CBP/metabolismo
16.
Kidney Int ; 89(5): 1125-1135, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27083286

RESUMEN

Many diabetic patients suffer from declining renal function without developing albuminuria. To identify alternative biomarkers for diabetic nephropathy (DN) we performed urinary peptidomic analysis in a rodent model in which hyperglycemia and hypertension synergize to promote renal pathologic changes consistent with human DN. We identified 297 increased and 15 decreased peptides in the urine of rats with DN compared with controls, including peptides derived from proteins associated with DN and novel candidate biomarkers. We confirmed by ELISA that one of the parent proteins, urinary epidermal growth factor (uEGF), was more than 2-fold reduced in rats with DN in comparison with controls. To assess the clinical utility of uEGF we examined renal outcomes in 642 participants from the Edinburgh Type 2 Diabetes Study who were normoalbuminuric and had preserved renal function at baseline. After adjustment for established renal risk factors, a lower uEGF to creatinine ratio was associated with new-onset estimated glomerular filtration rate less than 60 ml/min per 1.73m(2) (odds ratio 0.48; 95% confidence interval, 0.26-0.90), rapid (over 5% per annum) decline in renal function (odds ratio 0.44; 95% confidence interval, 0.27-0.72) or the composite of both outcomes (odds ratio 0.38; 95% confidence interval, 0.24-0.62). Thus, the utility of a low uEGF to creatinine ratio as a biomarker of progressive decline in renal function in normoalbuminuric patients should be assessed in additional populations.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/orina , Receptores ErbB/orina , Proteinuria/orina , Proteómica , Receptor ErbB-2/orina , Anciano , Animales , Biomarcadores/orina , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Creatinina/orina , Diabetes Mellitus Tipo 2/diagnóstico , Nefropatías Diabéticas/diagnóstico , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Tasa de Filtración Glomerular , Humanos , Hipertensión/complicaciones , Estimación de Kaplan-Meier , Riñón/fisiopatología , Modelos Logísticos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Análisis Multivariante , Oportunidad Relativa , Valor Predictivo de las Pruebas , Proteómica/métodos , Ratas Transgénicas , Factores de Riesgo , Escocia , Urinálisis
17.
Biochem J ; 469(1): 25-32, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25891661

RESUMEN

Protein S-glutathionylation is a reversible post-translational modification regulating sulfhydryl homeostasis. However, little is known about the proteins and pathways regulated by S-glutathionylation in whole organisms and current approaches lack the sensitivity to examine this modification under basal conditions. We now report the quantification and identification of S-glutathionylated proteins from animal tissue, using a highly sensitive methodology combining high-accuracy proteomics with tandem mass tagging to provide precise, extensive coverage of S-glutathionylated targets in mouse liver. Critically, we show significant enrichment of S-glutathionylated mitochondrial and Krebs cycle proteins, identifying that S-glutathionylation is heavily involved in energy metabolism processes in vivo. Furthermore, using mice nulled for GST Pi (GSTP) we address the potential for S-glutathionylation to be mediated enzymatically. The data demonstrate the impact of S-glutathionylation in cellular homeostasis, particularly in relation to energy regulation and is of significant interest for those wishing to examine S-glutathionylation in an animal model.


Asunto(s)
Glutatión Transferasa/metabolismo , Glutatión/metabolismo , Hígado/metabolismo , Proteínas Mitocondriales/metabolismo , Procesamiento Proteico-Postraduccional , Proteoma/metabolismo , Animales , Glutatión/genética , Glutatión Transferasa/genética , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteoma/genética
18.
J Neurosci ; 34(38): 12904-18, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25232125

RESUMEN

Postnatal synapse elimination plays a critical role in sculpting and refining neural connectivity throughout the central and peripheral nervous systems, including the removal of supernumerary axonal inputs from neuromuscular junctions (NMJs). Here, we reveal a novel and important role for myelinating glia in regulating synapse elimination at the mouse NMJ, where loss of a single glial cell protein, the glial isoform of neurofascin (Nfasc155), was sufficient to disrupt postnatal remodeling of synaptic circuitry. Neuromuscular synapses were formed normally in mice lacking Nfasc155, including the establishment of robust neuromuscular synaptic transmission. However, loss of Nfasc155 was sufficient to cause a robust delay in postnatal synapse elimination at the NMJ across all muscle groups examined. Nfasc155 regulated neuronal remodeling independently of its canonical role in forming paranodal axo-glial junctions, as synapse elimination occurred normally in mice lacking the axonal paranodal protein Caspr. Rather, high-resolution proteomic screens revealed that loss of Nfasc155 from glial cells was sufficient to disrupt neuronal cytoskeletal organization and trafficking pathways, resulting in reduced levels of neurofilament light (NF-L) protein in distal axons and motor nerve terminals. Mice lacking NF-L recapitulated the delayed synapse elimination phenotype observed in mice lacking Nfasc155, suggesting that glial cells regulate synapse elimination, at least in part, through modulation of the axonal cytoskeleton. Together, our study reveals a glial cell-dependent pathway regulating the sculpting of neuronal connectivity and synaptic circuitry in the peripheral nervous system.


Asunto(s)
Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/fisiología , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/fisiología , Unión Neuromuscular/fisiología , Sinapsis/fisiología , Animales , Axones/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/fisiología , Citoesqueleto/metabolismo , Ratones , Ratones Noqueados , Placa Motora/crecimiento & desarrollo , Neuronas Motoras/metabolismo , Factores de Crecimiento Nervioso/genética , Conducción Nerviosa/genética , Conducción Nerviosa/fisiología , Proteínas de Neurofilamentos/metabolismo , Neuroglía/metabolismo , Unión Neuromuscular/crecimiento & desarrollo , Isoformas de Proteínas/genética , Proteómica , Células de Schwann/metabolismo , Sinapsis/genética , Transmisión Sináptica/fisiología
19.
BMC Cancer ; 15: 885, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26555036

RESUMEN

BACKGROUND: Cyclin-dependent protein kinase-5 (CDK5) is an unusual member of the CDK family as it is not cell cycle regulated. However many of its substrates have roles in cell growth and oncogenesis, raising the possibility that CDK5 modulation could have therapeutic benefit. In order to establish whether changes in CDK5 activity are associated with oncogenesis one could quantify phosphorylation of CDK5 targets in disease tissue in comparison to appropriate controls. However the identity of physiological and pathophysiological CDK5 substrates remains the subject of debate, making the choice of CDK5 activity biomarkers difficult. METHODS: Here we use in vitro and in cell phosphorylation assays to identify novel features of CDK5 target sequence determinants that confer enhanced CDK5 selectivity, providing means to select substrate biomarkers of CDK5 activity with more confidence. We then characterize tools for the best CDK5 substrate we identified to monitor its phosphorylation in human tissue and use these to interrogate human tumour arrays. RESULTS: The close proximity of Arg/Lys amino acids and a proline two residues N-terminal to the phosphorylated residue both improve recognition of the substrate by CDK5. In contrast the presence of a proline two residues C-terminal to the target residue dramatically reduces phosphorylation rate. Serine-522 of Collapsin Response Mediator-2 (CRMP2) is a validated CDK5 substrate with many of these structural criteria. We generate and characterise phosphospecific antibodies to Ser522 and show that phosphorylation appears in human tumours (lung, breast, and lymphoma) in stark contrast to surrounding non-neoplastic tissue. In lung cancer the anti-phospho-Ser522 signal is positive in squamous cell carcinoma more frequently than adenocarcinoma. Finally we demonstrate that it is a specific and unusual splice variant of CRMP2 (CRMP2A) that is phosphorylated in tumour cells. CONCLUSIONS: For the first time this data associates altered CDK5 substrate phosphorylation with oncogenesis in some but not all tumour types, implicating altered CDK5 activity in aspects of pathogenesis. These data identify a novel oncogenic mechanism where CDK5 activation induces CRMP2A phosphorylation in the nuclei of tumour cells.


Asunto(s)
Carcinogénesis/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/genética , Proteínas del Tejido Nervioso/metabolismo , Secuencia de Aminoácidos , Biomarcadores de Tumor , Quinasa 5 Dependiente de la Ciclina/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas del Tejido Nervioso/genética , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Empalme del ARN/genética , Serina/metabolismo
20.
PLoS Genet ; 8(8): e1002936, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22952455

RESUMEN

Degeneration of synaptic and axonal compartments of neurons is an early event contributing to the pathogenesis of many neurodegenerative diseases, but the underlying molecular mechanisms remain unclear. Here, we demonstrate the effectiveness of a novel "top-down" approach for identifying proteins and functional pathways regulating neurodegeneration in distal compartments of neurons. A series of comparative quantitative proteomic screens on synapse-enriched fractions isolated from the mouse brain following injury identified dynamic perturbations occurring within the proteome during both initiation and onset phases of degeneration. In silico analyses highlighted significant clustering of proteins contributing to functional pathways regulating synaptic transmission and neurite development. Molecular markers of degeneration were conserved in injury and disease, with comparable responses observed in synapse-enriched fractions isolated from mouse models of Huntington's disease (HD) and spinocerebellar ataxia type 5. An initial screen targeting thirteen degeneration-associated proteins using mutant Drosophila lines revealed six potential regulators of synaptic and axonal degeneration in vivo. Mutations in CALB2, ROCK2, DNAJC5/CSP, and HIBCH partially delayed injury-induced neurodegeneration. Conversely, mutations in DNAJC6 and ALDHA1 led to spontaneous degeneration of distal axons and synapses. A more detailed genetic analysis of DNAJC5/CSP mutants confirmed that loss of DNAJC5/CSP was neuroprotective, robustly delaying degeneration in axonal and synaptic compartments. Our study has identified conserved molecular responses occurring within synapse-enriched fractions of the mouse brain during the early stages of neurodegeneration, focused on functional networks modulating synaptic transmission and incorporating molecular chaperones, cytoskeletal modifiers, and calcium-binding proteins. We propose that the proteins and functional pathways identified in the current study represent attractive targets for developing therapeutics aimed at modulating synaptic and axonal stability and neurodegeneration in vivo.


Asunto(s)
Lesiones Encefálicas , Drosophila , Degeneración Nerviosa , Sinapsis , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Animales , Axones/metabolismo , Axones/patología , Axones/fisiología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Calbindina 2 , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Ratones , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutación , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Proteómica , Proteína G de Unión al Calcio S100/genética , Proteína G de Unión al Calcio S100/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Tioléster Hidrolasas/genética , Tioléster Hidrolasas/metabolismo , Degeneración Walleriana/metabolismo , Degeneración Walleriana/patología , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA