Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 20(1): 413, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36076207

RESUMEN

BACKGROUND: Next generation sequencing (NGS) of human specimen is expected to improve prognosis and diagnosis of human diseases, but its sensitivity urges for well-defined sampling and standardized protocols in order to avoid error-prone conclusions. METHODS: In this study, large volumes of pooled human cerebrospinal fluid (CSF) were used to prepare RNA from human CSF-derived extracellular vesicles (EV) and from whole CSF, as well as from whole human serum and serum-derived EV. In all four fractions small and long coding and non-coding RNA expression was analyzed with NGS and transcriptome analyses. RESULTS: We show, that the source of sampling has a large impact on the acquired NGS pattern, and differences between small RNA fractions are more distinct than differences between long RNA fractions. The highest percentual discrepancy between small RNA fractions and the second highest difference between long RNA fractions is seen in the comparison of CSF-derived EV and whole CSF. Differences between miR (microRNA) and mRNA fractions of EV and the respective whole body fluid have the potential to affect different cellular and biological processes. I.e. a comparison of miR in both CSF fractions reveals that miR from EV target four transcripts sets involved in neurobiological processes, whereas eight others, also involved in neurobiological processes are targeted by miR found in whole CSF only. Likewise, three mRNAs sets derived from CSF-derived EV are associated with neurobiological and six sets with mitochondrial metabolism, whereas no such mRNA transcript sets are found in the whole CSF fraction. We show that trace amounts of blood-derived contaminations of CSF can bias RNA-based CSF diagnostics. CONCLUSIONS: This study shows that the composition of small and long RNA differ significantly between whole body fluid and its respective EV fraction and thus can affect different cellular and molecular functions. Trace amounts of blood-derived contaminations of CSF can bias CSF analysis. This has to be considered for a meaningful RNA-based diagnostics. Our data imply a transport of EV from serum to CSF across the blood-brain barrier.


Asunto(s)
Fenómenos Biológicos , Vesículas Extracelulares , MicroARNs , Vesículas Extracelulares/genética , Humanos , MicroARNs/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcriptoma/genética
2.
PLoS Pathog ; 14(1): e1006802, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29357384

RESUMEN

Increasing evidence indicates that microRNAs (miRNAs) are contributing factors to neurodegeneration. Alterations in miRNA signatures have been reported in several neurodegenerative dementias, but data in prion diseases are restricted to ex vivo and animal models. The present study identified significant miRNA expression pattern alterations in the frontal cortex and cerebellum of sporadic Creutzfeldt-Jakob disease (sCJD) patients. These changes display a highly regional and disease subtype-dependent regulation that correlates with brain pathology. We demonstrate that selected miRNAs are enriched in sCJD isolated Argonaute(Ago)-binding complexes in disease, indicating their incorporation into RNA-induced silencing complexes, and further suggesting their contribution to disease-associated gene expression changes. Alterations in the miRNA-mRNA regulatory machinery and perturbed levels of miRNA biogenesis key components in sCJD brain samples reported here further implicate miRNAs in sCJD gene expression (de)regulation. We also show that a subset of sCJD-altered miRNAs are commonly changed in Alzheimer's disease, dementia with Lewy bodies and fatal familial insomnia, suggesting potential common mechanisms underlying these neurodegenerative processes. Additionally, we report no correlation between brain and cerebrospinal fluid (CSF) miRNA-profiles in sCJD, indicating that CSF-miRNA profiles do not faithfully mirror miRNA alterations detected in brain tissue of human prion diseases. Finally, utilizing a sCJD MM1 mouse model, we analyzed the miRNA deregulation patterns observed in sCJD in a temporal manner. While fourteen sCJD-related miRNAs were validated at clinical stages, only two of those were changed at early symptomatic phase, suggesting that the miRNAs altered in sCJD may contribute to later pathogenic processes. Altogether, the present work identifies alterations in the miRNA network, biogenesis and miRNA-mRNA silencing machinery in sCJD, whereby contributions to disease mechanisms deserve further investigation.


Asunto(s)
Síndrome de Creutzfeldt-Jakob/clasificación , Síndrome de Creutzfeldt-Jakob/genética , MicroARNs/genética , Interferencia de ARN , Transcriptoma , Adulto , Anciano , Anciano de 80 o más Años , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Casos y Controles , Síndrome de Creutzfeldt-Jakob/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , MicroARNs/biosíntesis , Persona de Mediana Edad
3.
J Neuroinflammation ; 15(1): 175, 2018 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880000

RESUMEN

BACKGROUND: Bacterial meningitis is associated with high mortality and long-term neurological sequelae. Increasing the phagocytic activity of microglia could improve the resistance of the CNS against infections. We studied the influence of activin A, a member of the TGF-ß family with known immunoregulatory and neuroprotective effects, on the functions of microglial cells in vitro. METHODS: Primary murine microglial cells were treated with activin A (0.13 ng/ml-13 µg/ml) alone or in combination with agonists of TLR2, 4, and 9. Phagocytosis of Escherichia coli K1 as well as release of TNF-α, IL-6, CXCL1, and NO was assessed. RESULTS: Activin A dose-dependently enhanced the phagocytosis of Escherichia coli K1 by microglial cells activated by agonists of TLR2, 4, and 9 without further increasing NO and proinflammatory cytokine release. Cell viability of microglial cells was not affected by activin A. CONCLUSIONS: Priming of microglial cells with activin A could increase the elimination of bacteria in bacterial CNS infections. This preventive strategy could improve the resistance of the brain to infections, particularly in elderly and immunocompromised patients.


Asunto(s)
Activinas/farmacología , Citocinas/metabolismo , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Receptores Toll-Like/agonistas , Animales , Animales Recién Nacidos , Encéfalo/citología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Escherichia coli/fisiología , Humanos , Recién Nacido , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Lectinas de Plantas/metabolismo , Receptores Toll-Like/metabolismo
4.
J Neurochem ; 134(2): 261-75, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25807858

RESUMEN

A lesion to the rat rubrospinal tract is a model for traumatic spinal cord lesions and results in atrophy of the red nucleus neurons, axonal dieback, and locomotor deficits. In this study, we used adeno-associated virus (AAV)-mediated over-expression of BAG1 and ROCK2-shRNA in the red nucleus to trace [by co-expression of enhanced green fluorescent protein (EGFP)] and treat the rubrospinal tract after unilateral dorsal hemisection. We investigated the effects of targeted gene therapy on neuronal survival, axonal sprouting of the rubrospinal tract, and motor recovery 12 weeks after unilateral dorsal hemisection at Th8 in rats. In addition to the evaluation of BAG1 and ROCK2 as therapeutic targets in spinal cord injury, we aimed to demonstrate the feasibility and the limits of an AAV-mediated protein over-expression versus AAV.shRNA-mediated down-regulation in this traumatic CNS lesion model. Our results demonstrate that BAG1 and ROCK2-shRNA both promote neuronal survival of red nucleus neurons and enhance axonal sprouting proximal to the lesion.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Regeneración Nerviosa/fisiología , Neuronas/patología , Traumatismos de la Médula Espinal/patología , Factores de Transcripción/biosíntesis , Quinasas Asociadas a rho/biosíntesis , Animales , Axones , Secuencia de Bases , Western Blotting , Supervivencia Celular , Proteínas de Unión al ADN/genética , Dependovirus , Modelos Animales de Enfermedad , Femenino , Terapia Genética/métodos , Vectores Genéticos , Inmunohistoquímica , Datos de Secuencia Molecular , ARN Interferente Pequeño , Ratas , Ratas Wistar , Recuperación de la Función , Núcleo Rojo/patología , Factores de Transcripción/genética , Quinasas Asociadas a rho/genética
5.
Neurobiol Dis ; 73: 150-62, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25283984

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder with prominent neuronal cell death in the substantia nigra (SN) and other parts of the brain. Previous studies in models of traumatic and neurodegenerative CNS disease showed that pharmacological inhibition of Rho-associated kinase (ROCK), a molecule involved in inhibitory signaling in the CNS, by small-molecule inhibitors improves neuronal survival and increases regeneration. Most small-molecule inhibitors, however, offer only limited target specificity and also inhibit other kinases, including both ROCK isoforms. To establish the role of the predominantly brain-expressed ROCK2 isoform in models of regeneration and PD, we used adeno-associated viral vectors (AAV) to specifically knockdown ROCK2 in neurons. Rat primary midbrain neurons (PMN) were transduced with AAV expressing short-hairpin-RNA (shRNA) against ROCK2 and LIM-domain kinase 1 (LIMK1), one of the downstream targets of ROCK2. While knock-down of ROCK2 and LIMK1 both enhanced neurite regeneration in a traumatic scratch lesion model, only ROCK2-shRNA protected PMN against 1-methyl-4-phenylpyridinium (MPP+) toxicity. Moreover, AAV.ROCK2-shRNA increased levels of the pro-survival markers Bcl-2 and phospho-Erk1. In vivo, AAV.ROCK2-shRNA vectors were injected into the ipsilateral SN and a unilateral 6-OHDA striatal lesion was performed. After four weeks, behavioral, immunohistochemical and biochemical alterations were investigated. Downregulation of ROCK2 protected dopaminergic neurons in the SN from 6-OHDA-induced degeneration and resulted in significantly increased TH-positive neuron numbers. This effect, however, was confined to nigral neuronal somata as striatal terminal density, dopamine and metabolite levels were not significantly preserved. Interestingly, motor behavior was improved in the ROCK2-shRNA treated animals compared to control after four weeks. Our studies thus confirm ROCK2 as a promising therapeutic target in models of PD and demonstrate that neuron-specific inhibition of ROCK2 promotes survival of lesioned dopaminergic neurons.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Regulación hacia Abajo/fisiología , Degeneración Nerviosa/etiología , Degeneración Nerviosa/patología , Enfermedad de Parkinson/complicaciones , Quinasas Asociadas a rho/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Adrenérgicos/toxicidad , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Vectores Genéticos/fisiología , Ácido Homovanílico , Quinasas Lim/genética , Quinasas Lim/metabolismo , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Desempeño Psicomotor , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Sustancia Negra/patología , Tirosina 3-Monooxigenasa , Quinasas Asociadas a rho/genética
6.
Acta Neuropathol Commun ; 12(1): 82, 2024 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-38812004

RESUMEN

Neurons pose a particular challenge to degradative processes like autophagy due to their long and thin processes. Autophagic vesicles (AVs) are formed at the tip of the axon and transported back to the soma. This transport is essential since the final degradation of the vesicular content occurs only close to or in the soma. Here, we established an in vivo live-imaging model in the rat optic nerve using viral vector mediated LC3-labeling and two-photon-microscopy to analyze axonal transport of AVs. Under basal conditions in vivo, 50% of the AVs are moving with a majority of 85% being transported in the retrograde direction. Transport velocity is higher in the retrograde than in the anterograde direction. A crush lesion of the optic nerve results in a rapid breakdown of retrograde axonal transport while the anterograde transport stays intact over several hours. Close to the lesion site, the formation of AVs is upregulated within the first 6 h after crush, but the clearance of AVs and the levels of lysosomal markers in the adjacent axon are reduced. Expression of p150Glued, an adaptor protein of dynein, is significantly reduced after crush lesion. In vitro, fusion and colocalization of the lysosomal marker cathepsin D with AVs are reduced after axotomy. Taken together, we present here the first in vivo analysis of axonal AV transport in the mammalian CNS using live-imaging. We find that axotomy leads to severe defects of retrograde motility and a decreased clearance of AVs via the lysosomal system.


Asunto(s)
Autofagia , Transporte Axonal , Nervio Óptico , Animales , Transporte Axonal/fisiología , Nervio Óptico/patología , Nervio Óptico/metabolismo , Ratas , Autofagia/fisiología , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Masculino , Axones/patología , Axones/metabolismo , Degeneración Nerviosa/patología , Degeneración Nerviosa/metabolismo , Ratas Sprague-Dawley , Femenino
7.
Neurobiol Dis ; 51: 168-76, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23174179

RESUMEN

The ability of fish retinal ganglion cells (RGCs) to regenerate their axons was shown to require the re-expression and function of the two proteins reggie-1 and -2. RGCs in mammals fail to upregulate reggie expression and to regenerate axons after lesion suggesting the possibility that induced upregulation might promote regeneration. In the present study, RGCs in adult rats were induced to express reggie-1 by intravitreal injection of adeno-associated viral vectors (AAV2/1) expressing reggie-1 (AAV.R1-EGFP) 14d prior to optic nerve crush. Four weeks later, GAP-43-positive regenerating axons had crossed the lesion and grown into the nerve at significantly higher numbers and length (up to 5mm) than the control transduced with AAV.EGFP. Consistently, after transduction with AAV.R1-EGFP as opposed to AAV.EGFP, primary RGCs in vitro grew long axons on chondroitin sulfate proteoglycan (CSPG) and Nogo-A, both glial cell-derived inhibitors of neurite growth, suggesting that reggie-1 can provide neurons with the ability to override inhibitors of neurite growth. This reggie-1-mediated enhancement of growth was reproduced in mouse hippocampal and N2a neurons which generated axons 40-60% longer than their control counterparts. This correlates with the reggie-1-dependent activation of Src and PI3 kinase (PI3K), of the Rho family GTPase Rac1 and downstream effectors such as cofilin. This increased growth also depends on TC10, the GTPase involved in cargo delivery to the growth cone. Thus, the upregulation of reggie-1 in mammalian neurons provides nerve cells with neuron-intrinsic properties required for axon growth and successful regeneration in the adult mammalian CNS.


Asunto(s)
Axones/metabolismo , Proteínas de la Membrana/biosíntesis , Regeneración Nerviosa/fisiología , Neuritas/metabolismo , Nervio Óptico/metabolismo , Animales , Western Blotting , Ratones , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Transducción Genética , Regulación hacia Arriba
8.
Acta Neuropathol ; 126(2): 251-65, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23754622

RESUMEN

MicroRNAs (miRNAs) are highly conserved non-coding RNAs modulating gene expression via mRNA binding. Recent work suggests an involvement of miRNAs in cardiovascular diseases including stroke. As such, the brain-abundant miR-124 and its transcriptional repressor RE1-silencing transcription factor (REST) do not only have elementary roles in the developing and the adult brain, but also alter expression upon cerebral ischemia. However, the therapeutic potential of miR-124 against stroke and the mechanisms involved remain elusive. Here, we analyzed the therapeutic potential of ectopic miR-124 against stroke and its underlying mechanisms with regard to the interaction between miR-124 and REST. Our results show that viral vector-mediated miR-124 delivery increased the resistance of cultured oxygen-glucose-deprived cortical neurons in vitro and reduced brain injury as well as functional impairment in mice submitted to middle cerebral artery occlusion. Likewise, miR-124 induced enhanced neurovascular remodeling leading to increased angioneurogenesis 8 weeks post-stroke. While REST abundance increased upon stroke, the increase was prevented by miR-124 despite a so far unknown negative feedback loop between miR-124 and REST. Rather, miR-124 decreased the expression of the deubiquitinating enzyme Usp14, which has two conserved miR-124-binding sites in the 3'UTR of its mRNA, and thereby mediated reduced REST levels. The down-regulation of REST by miR-124 was also mimicked by the Usp14 inhibitor IU-1, suggesting that miR-124 promotes neuronal survival under ischemic conditions via Usp14-dependent REST degradation. Ectopic miR-124 expression, therefore, appears as an attractive and novel tool in stroke treatment, mediating neuroprotection via a hitherto unknown mechanism that involves Usp14-dependent REST degradation.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/patología , MicroARNs/fisiología , Neuronas/patología , Proteínas Represoras/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Isquemia Encefálica/metabolismo , Calpaína/metabolismo , Supervivencia Celular/fisiología , Glucosa/farmacología , Células HEK293 , Humanos , Peroxidación de Lípido/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Oxígeno/farmacología , Receptores AMPA/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Ubiquitinación/fisiología
9.
Proc Natl Acad Sci U S A ; 107(13): 6064-9, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20231460

RESUMEN

Axonal degeneration is an initial key step in traumatic and neurodegenerative CNS disorders. We established a unique in vivo epifluorescence imaging paradigm to characterize very early events in axonal degeneration in the rat optic nerve. Single retinal ganglion cell axons were visualized by AAV-mediated expression of dsRed and this allowed the quantification of postlesional acute axonal degeneration (AAD). EM analysis revealed severe structural alterations of the cytoskeleton, cytoplasmatic vacuolization, and the appearance of autophagosomes within the first hours after lesion. Inhibition of autophagy resulted in an attenuation of acute axonal degeneration. Furthermore, a rapid increase of intraaxonal calcium levels following crush lesion could be visualized using a calcium-sensitive dye. Application of calcium channel inhibitors prevented crush-induced calcium increase and markedly attenuated axonal degeneration, whereas application of a calcium ionophore aggravated the degenerative phenotype. We finally demonstrate that increased postlesional autophagy is calcium dependent and thus mechanistically link autophagy and intraaxonal calcium levels. Both processes are proposed to be major targets for the manipulation of axonal degeneration in future therapeutic settings.


Asunto(s)
Axones/patología , Degeneración Nerviosa/patología , Nervio Óptico/patología , Animales , Autofagia , Señalización del Calcio , Dependovirus/genética , Femenino , Vectores Genéticos , Proteínas Luminiscentes/genética , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Degeneración Nerviosa/fisiopatología , Nervio Óptico/fisiopatología , Traumatismos del Nervio Óptico/patología , Traumatismos del Nervio Óptico/fisiopatología , Ratas , Ratas Wistar , Proteínas Recombinantes/genética , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/fisiología , Factores de Tiempo
10.
Sci Rep ; 12(1): 14387, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35999340

RESUMEN

The axonal cytoskeleton is organized in a highly periodic structure, the membrane-associated periodic skeleton (MPS), which is essential to maintain the structure and function of the axon. Here, we use stimulated emission depletion microscopy of primary rat cortical neurons in microfluidic chambers to analyze the temporal and spatial sequence of MPS formation at the distal end of growing axons and during regeneration after axotomy. We demonstrate that the MPS does not extend continuously into the growing axon but develops from patches of periodic ßII-spectrin arrangements that grow and coalesce into a continuous scaffold. We estimate that the underlying sequence of assembly, elongation, and subsequent coalescence of periodic ßII-spectrin patches takes around 15 h. Strikingly, we find that development of the MPS occurs faster in regenerating axons after axotomy and note marked differences in the morphology of the growth cone and adjacent axonal regions between regenerating and unlesioned axons. Moreover, we find that inhibition of the spectrin-cleaving enzyme calpain accelerates MPS formation in regenerating axons and increases the number of regenerating axons after axotomy. Taken together, we provide here a detailed nanoscale analysis of MPS development in growing axons.


Asunto(s)
Citoesqueleto , Espectrina , Animales , Axones/fisiología , Conos de Crecimiento , Regeneración Nerviosa/fisiología , Ratas , Regeneración
11.
Cell Death Dis ; 12(2): 213, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637688

RESUMEN

Axonal damage is an early step in traumatic and neurodegenerative disorders of the central nervous system (CNS). Damaged axons are not able to regenerate sufficiently in the adult mammalian CNS, leading to permanent neurological deficits. Recently, we showed that inhibition of the autophagic protein ULK1 promotes neuroprotection in different models of neurodegeneration. Moreover, we demonstrated previously that axonal protection improves regeneration of lesioned axons. However, whether axonal protection mediated by ULK1 inhibition could also improve axonal regeneration is unknown. Here, we used an adeno-associated viral (AAV) vector to express a dominant-negative form of ULK1 (AAV.ULK1.DN) and investigated its effects on axonal regeneration in the CNS. We show that AAV.ULK1.DN fosters axonal regeneration and enhances neurite outgrowth in vitro. In addition, AAV.ULK1.DN increases neuronal survival and enhances axonal regeneration after optic nerve lesion, and promotes long-term axonal protection after spinal cord injury (SCI) in vivo. Interestingly, AAV.ULK1.DN also increases serotonergic and dopaminergic axon sprouting after SCI. Mechanistically, AAV.ULK1.DN leads to increased ERK1 activation and reduced expression of RhoA and ROCK2. Our findings outline ULK1 as a key regulator of axonal degeneration and regeneration, and define ULK1 as a promising target to promote neuroprotection and regeneration in the CNS.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Axones/metabolismo , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Regeneración Nerviosa , Traumatismos del Nervio Óptico/terapia , Nervio Óptico/metabolismo , Traumatismos de la Médula Espinal/terapia , Médula Espinal/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Axones/patología , Células Cultivadas , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Regulación hacia Abajo , Femenino , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proyección Neuronal , Nervio Óptico/patología , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Ratas Wistar , Neuronas Serotoninérgicas/metabolismo , Neuronas Serotoninérgicas/patología , Médula Espinal/patología , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Factores de Tiempo , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
12.
Neurobiol Dis ; 38(3): 395-404, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20211260

RESUMEN

Malfunction of the ubiquitin-proteasome system has been implicated as a causal factor in the pathogenesis of aggregation-related disorders, e.g. Parkinson's disease. We show here that Transforming growth factor-beta 1 (TGF-beta), a multifunctional cytokine and trophic factor for dopaminergic (DAergic) neurons modulates proteasome function in primary midbrain neurons. TGF-beta differentially inhibited proteasomal subactivities with a most pronounced time-dependent inhibition of the peptidyl-glutamyl peptide hydrolyzing-like and chymotrypsin-like subactivity. Regulation of proteasomal activity could be specifically quantified in the DAergic subpopulation. Protein blot analysis revealed an accumulation of ubiquitinated proteins after TGF-beta treatment. The identity of these enriched proteins was further analyzed by 2D-gel electrophoresis and mass spectrometry. We found epidermal fatty acid binding protein (EFABP) to be strongly increased and ubiquitinated after TGF-beta treatment and confirmed this finding by co-immunoprecipitation. While application of TGF-beta increased neurite regeneration in a scratch lesion model, downregulation of EFABP by siRNA significantly decreased this effect. We thus postulate that a differential regulation of proteasomal function, as demonstrated for TGF-beta, can result in an enrichment of proteins, such as EFABP, that mediate physiological functions, such as neurite regeneration.


Asunto(s)
Proteínas del Ojo/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Aumento de la Célula , Células Cultivadas , Dopamina/metabolismo , Hidrólisis , Mesencéfalo/fisiología , Regeneración Nerviosa/fisiología , Neuronas/fisiología , Ratas , Ratas Wistar , Factores de Tiempo , Ubiquitinación
13.
Mol Neurobiol ; 57(2): 685-697, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31446549

RESUMEN

Loss of nigrostriatal projections by axonal degeneration is a key early event in Parkinson's disease (PD) pathophysiology, being accountable for the lack of dopamine in the nigrostriatal system and resulting in motor symptoms such as bradykinesia, rigidity, and tremor. Since autophagy is an important mechanism contributing to axonal degeneration, we aimed to evaluate the effects of competitive autophagy inhibition in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD in vivo. Adeno-associated viral vector (AAV)-mediated overexpression of a dominant-negative form of the unc-51 like autophagy-initiating kinase (ULK1.DN) in the substantia nigra was induced 3 weeks before MPTP treatment. Analysis of motor behavior demonstrated a significant improvement of ULK1.DN expressing mice after MPTP treatment. Immunohistochemical analyses of dopaminergic nigral neurons and nigrostriatal projections revealed a significant protection from MPTP-induced neurotoxicity after ULK1.DN expression. Western blot analysis linked these findings to an activation of mTOR signaling. Taken together, our results indicate that expression of ULK1.DN can attenuate MPTP-induced axonal neurodegeneration, suggesting that ULK1 could be a promising novel target in the treatment of PD.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Dependovirus/metabolismo , Genes Dominantes , Actividad Motora , Neuronas/enzimología , Neuronas/patología , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/fisiopatología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Conducta Animal , Supervivencia Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Células HEK293 , Humanos , Masculino , Metaboloma , Ratones Endogámicos C57BL , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/metabolismo
14.
Cell Death Differ ; 27(10): 2810-2827, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32341448

RESUMEN

Axonal degeneration is a key and early pathological feature in traumatic and neurodegenerative disorders of the CNS. Following a focal lesion to axons, extended axonal disintegration by acute axonal degeneration (AAD) occurs within several hours. During AAD, the accumulation of autophagic proteins including Unc-51 like autophagy activating kinase 1 (ULK1) has been demonstrated, but its role is incompletely understood. Here, we study the effect of ULK1 inhibition in different models of lesion-induced axonal degeneration in vitro and in vivo. Overexpression of a dominant negative of ULK1 (ULK1.DN) in primary rat cortical neurons attenuates axotomy-induced AAD in vitro. Both ULK1.DN and the ULK1 inhibitor SBI-0206965 protect against AAD after rat optic nerve crush in vivo. ULK1.DN additionally attenuates long-term axonal degeneration after rat spinal cord injury in vivo. Mechanistically, ULK1.DN decreases autophagy and leads to an mTOR-mediated increase in translational proteins. Consistently, treatment with SBI-0206965 results in enhanced mTOR activation. ULK1.DN additionally modulates the differential splicing of the degeneration-associated genes Kif1b and Ddit3. These findings uncover ULK1 as an important mediator of axonal degeneration in vitro and in vivo, and elucidate its function in splicing, defining it as a putative therapeutic target.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia , Axones , Sistema Nervioso Central , Degeneración Nerviosa , Enfermedades Neurodegenerativas , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Homólogo de la Proteína 1 Relacionada con la Autofagia/fisiología , Axones/metabolismo , Axones/patología , Células Cultivadas , Sistema Nervioso Central/lesiones , Sistema Nervioso Central/metabolismo , Femenino , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Cultivo Primario de Células , Ratas
15.
Neurosci Lett ; 413(3): 241-4, 2007 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-17194540

RESUMEN

Activin A levels are elevated in the cerebrospinal fluid (CSF) of patients with meningitis and in the sera of patients with sepsis. The source(s) of the elevated concentrations of activin A in CSF and serum have not yet been discovered. Here we demonstrate that primary mouse microglial cells and peritoneal macrophages release activin A after treatment with agonists of Toll-like receptor (TLR) 2, 4, and 9. These findings provide further evidence for a role of activin in the innate immune response and suggest that microglial cells and macrophages are a source of elevated activin A concentrations observed in the CSF during bacterial meningitis and in the systemic circulation during sepsis.


Asunto(s)
Activinas/metabolismo , Macrófagos Peritoneales/metabolismo , Microglía/metabolismo , Receptores Toll-Like/agonistas , Animales , Animales Recién Nacidos , Encéfalo/citología , Células Cultivadas , Ciclopropanos/farmacología , Guanosina/análogos & derivados , Guanosina/farmacología , Lipopolisacáridos/farmacología , Lipoproteínas/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Receptores Toll-Like/fisiología
16.
Mol Neurobiol ; 54(1): 72-86, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26732591

RESUMEN

Axonal degeneration is one of the initial steps in many traumatic and neurodegenerative central nervous system (CNS) disorders and thus a promising therapeutic target. A focal axonal lesion is followed by acute axonal degeneration (AAD) of both adjacent axon parts, before proximal and distal parts follow different degenerative fates at later time points. Blocking calcium influx by calcium channel inhibitors was previously shown to attenuate AAD after optic nerve crush (ONC). However, it remains unclear whether the attenuation of AAD also promotes consecutive axonal regeneration. Here, we used a rat ONC model to study the effects of calcium channel inhibitors on axonal degeneration, retinal ganglion cell (RGC) survival, and axonal regeneration, as well as the molecular mechanisms involved. Application of calcium channel inhibitors attenuated AAD after ONC and preserved axonal integrity as visualized by live imaging of optic nerve axons. Consecutively, this resulted in improved survival of RGCs and improved axonal regeneration at 28 days after ONC. We show further that calcium channel inhibition attenuated lesion-induced calpain activation in the proximity of the crush and inhibited the activation of the c-Jun N-terminal kinase pathway. Pro-survival signaling via Akt in the retina was also increased. Our data thus show that attenuation of AAD improves consecutive neuronal survival and axonal regeneration and that calcium channel inhibitors could be valuable tools for therapeutic interventions in traumatic and degenerative CNS disorders.


Asunto(s)
Axones/fisiología , Bloqueadores de los Canales de Calcio/uso terapéutico , Regeneración Nerviosa/fisiología , Traumatismos del Nervio Óptico/prevención & control , Células Ganglionares de la Retina/fisiología , Animales , Axones/efectos de los fármacos , Axones/patología , Bloqueadores de los Canales de Calcio/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Femenino , Compresión Nerviosa , Regeneración Nerviosa/efectos de los fármacos , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Nervio Óptico/fisiología , Traumatismos del Nervio Óptico/patología , Ratas , Ratas Wistar , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología
17.
Acta Neuropathol Commun ; 5(1): 35, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28449707

RESUMEN

Sporadic Creutzfeldt-Jakob disease (sCJD) is the most prevalent form of human prion disease and it is characterized by the presence of neuronal loss, spongiform degeneration, chronic inflammation and the accumulation of misfolded and pathogenic prion protein (PrPSc). The molecular mechanisms underlying these alterations are largely unknown, but the presence of intracellular neuronal calcium (Ca2+) overload, a general feature in models of prion diseases, is suggested to play a key role in prion pathogenesis.Here we describe the presence of massive regulation of Ca2+ responsive genes in sCJD brain tissue, accompanied by two Ca2+-dependent processes: endoplasmic reticulum stress and the activation of the cysteine proteases Calpains 1/2. Pathogenic Calpain proteins activation in sCJD is linked to the cleavage of their cellular substrates, impaired autophagy and lysosomal damage, which is partially reversed by Calpain inhibition in a cellular prion model. Additionally, Calpain 1 treatment enhances seeding activity of PrPSc in a prion conversion assay. Neuronal lysosomal impairment caused by Calpain over activation leads to the release of the lysosomal protease Cathepsin S that in sCJD mainly localises in axons, although massive Cathepsin S overexpression is detected in microglial cells. Alterations in Ca2+ homeostasis and activation of Calpain-Cathepsin axis already occur at pre-clinical stages of the disease as detected in a humanized sCJD mouse model.Altogether our work indicates that unbalanced Calpain-Cathepsin activation is a relevant contributor to the pathogenesis of sCJD at multiple molecular levels and a potential target for therapeutic intervention.


Asunto(s)
Encéfalo/metabolismo , Calcio/metabolismo , Calpaína/metabolismo , Catepsinas/metabolismo , Síndrome de Creutzfeldt-Jakob/metabolismo , Homeostasis/fisiología , Animales , Encéfalo/patología , Cationes Bivalentes/metabolismo , Células Cultivadas , Síndrome de Creutzfeldt-Jakob/patología , Modelos Animales de Enfermedad , Humanos , Lisosomas/metabolismo , Lisosomas/patología , Mesocricetus , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Proteínas PrPSc/metabolismo , Ratas Wistar , Proteínas Recombinantes/metabolismo , Ovinos
18.
J Neurol Sci ; 250(1-2): 50-7, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16920154

RESUMEN

OBJECTIVE: Activin A, and its binding protein, follistatin (FS), are expressed in the central nervous system (CNS). We have previously shown elevated concentrations of FS in the cerebrospinal fluid (CSF) of patients with meningitis and increased concentrations of activin A in the CSF of rabbits with bacterial meningitis. METHODS: We measured CSF and serum concentrations of activin A and FS in normal subjects and in patients with various neurological diseases using previously validated immunoassays specific for activin A or FS. RESULTS: In healthy persons, serum concentrations of both activin A and FS were age-dependent. In CSF, concentrations of activin A ranged from 0.03 to 0.33 ng/ml and were strongly correlated with age in both sexes, whereas FS CSF concentrations were below the assay detection limit in most of the patients. Activin A concentrations in CSF of patients with various neurological diseases, including meningitis, chronic inflammatory CNS diseases, neurodegenerative diseases, tumors in the CNS, cerebral ischemia, intracerebral/subarachnoid hemorrhages, subdural hemorrhages and epileptic seizures, were compared with age- and sex-matched control patients. The comparisons revealed significantly elevated concentrations of activin A in patients with meningitis (P=0.017). Serum concentrations of activin A or FS were not affected by any of the neurological diseases examined. CONCLUSIONS: Our results show for the first time that in normal subjects concentrations of activin A in CSF are correlated with age, and furthermore, that activin A CSF concentrations are elevated in patients with meningitis. The latter underlines a role for activin A in acute inflammatory processes within the CNS.


Asunto(s)
Activinas/líquido cefalorraquídeo , Envejecimiento/metabolismo , Proteínas del Líquido Cefalorraquídeo/metabolismo , Meningitis/líquido cefalorraquídeo , Meningitis/diagnóstico , Activinas/análisis , Adulto , Anciano , Biomarcadores/líquido cefalorraquídeo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/microbiología , Sistema Nervioso Central/fisiopatología , Proteínas del Líquido Cefalorraquídeo/análisis , Encefalitis/líquido cefalorraquídeo , Encefalitis/diagnóstico , Encefalitis/fisiopatología , Femenino , Folistatina/análisis , Folistatina/líquido cefalorraquídeo , Humanos , Inmunoensayo , Masculino , Meningitis/fisiopatología , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Regulación hacia Arriba/fisiología
19.
Sci Rep ; 6: 37050, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27845394

RESUMEN

Axonal degeneration is a key initiating event in many neurological diseases. Focal lesions to axons result in a rapid disintegration of the perilesional axon by acute axonal degeneration (AAD) within several hours. However, the underlying molecular mechanisms of AAD are only incompletely understood. Here, we studied AAD in vivo through live-imaging of the rat optic nerve and in vitro in primary rat cortical neurons in microfluidic chambers. We found that calpain is activated early during AAD of the optic nerve and that calpain inhibition completely inhibits axonal fragmentation on the proximal side of the crush while it attenuates AAD on the distal side. A screening of calpain targets revealed that collapsin response mediator protein-2 (CRMP2) is a main downstream target of calpain activation in AAD. CRMP2-overexpression delayed bulb formation and rescued impairment of axonal mitochondrial transport after axotomy in vitro. In vivo, CRMP2-overexpression effectively protected the proximal axon from fragmentation within 6 hours after crush. Finally, a proteomic analysis of the optic nerve was performed at 6 hours after crush, which identified further proteins regulated during AAD, including several interactors of CRMP2. These findings reveal CRMP2 as an important mediator of AAD and define it as a putative therapeutic target.


Asunto(s)
Axones/metabolismo , Calpaína/metabolismo , Degeneración Nerviosa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades del Nervio Óptico/metabolismo , Enfermedad Aguda , Animales , Axones/patología , Células Cultivadas , Péptidos y Proteínas de Señalización Intercelular , Degeneración Nerviosa/patología , Enfermedades del Nervio Óptico/patología , Ratas
20.
Int Rev Cytol ; 218: 143-219, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12199517

RESUMEN

Non-coding ribonucleic acids (RNAs) do not contain a peptide-encoding open reading frame and are therefore not translated into proteins. They are expressed in all phyla, and in eukaryotic cells they are found in the nucleus, cytoplasm, and mitochondria. Non-coding RNAs either can exert structural functions, as do transfer and ribosomal RNAs, or they can regulate gene expression. Non-coding RNAs with regulatory functions differ in size ranging from a few nucleotides to over 100 kb and have diverse cell- or development-specific functions. Some of the non-coding RNAs associate with human diseases. This chapter summarizes the current knowledge about regulatory non-coding RNAs.


Asunto(s)
Células Eucariotas/metabolismo , Regulación de la Expresión Génica/genética , Genes Reguladores/genética , ARN no Traducido/genética , ARN no Traducido/metabolismo , Animales , Células Eucariotas/citología , Impresión Genómica/genética , Humanos , Células Procariotas/citología , Células Procariotas/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA