Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Annu Rev Cell Dev Biol ; 36: 165-189, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33021824

RESUMEN

As the world's population ages, neurodegenerative disorders are poised to become the commonest cause of death. Despite this, they remain essentially untreatable. Characterized pathologically both by the aggregation of disease-specific misfolded proteins and by changes in cellular stress responses, to date, therapeutic approaches have focused almost exclusively on reducing misfolded protein load-notably amyloid beta (Aß) in Alzheimer's disease. The repeated failure of clinical trials has led to despondency over the possibility that these disorders will ever be treated. We argue that this is in fact a time for optimism: Targeting various generic stress responses is emerging as an increasingly promising means of modifying disease progression across these disorders. New treatments are approaching clinical trials, while novel means of targeting aggregates could eventually act preventively in early disease.


Asunto(s)
Enfermedades Neurodegenerativas/terapia , Agregado de Proteínas , Estrés Fisiológico , Animales , Autofagosomas/metabolismo , Humanos , Lisosomas/metabolismo , Respuesta de Proteína Desplegada
2.
EMBO J ; 42(14): e113168, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37248947

RESUMEN

Enhanced expression of the cold-shock protein RNA binding motif 3 (RBM3) is highly neuroprotective both in vitro and in vivo. Whilst upstream signalling pathways leading to RBM3 expression have been described, the precise molecular mechanism of RBM3 cold induction remains elusive. To identify temperature-dependent modulators of RBM3, we performed a genome-wide CRISPR-Cas9 knockout screen using RBM3-reporter human iPSC-derived neurons. We found that RBM3 mRNA and protein levels are robustly regulated by several splicing factors, with heterogeneous nuclear ribonucleoprotein H1 (HNRNPH1) being the strongest positive regulator. Splicing analysis revealed that moderate hypothermia significantly represses the inclusion of a poison exon, which, when retained, targets the mRNA for nonsense-mediated decay. Importantly, we show that HNRNPH1 mediates this cold-dependent exon skipping via its thermosensitive interaction with a G-rich motif within the poison exon. Our study provides novel mechanistic insights into the regulation of RBM3 and provides further targets for neuroprotective therapeutic strategies.


Asunto(s)
Venenos , Humanos , Proteínas y Péptidos de Choque por Frío/metabolismo , Frío , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
3.
Mol Cell ; 73(3): 474-489.e5, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30595434

RESUMEN

Local translation is rapidly regulated by extrinsic signals during neural wiring, but its control mechanisms remain elusive. Here we show that the extracellular cue Sema3A induces an initial burst in local translation that precisely controls phosphorylation of the translation initiation factor eIF2α via the unfolded protein response (UPR) kinase PERK. Strikingly, in contrast to canonical UPR signaling, Sema3A-induced eIF2α phosphorylation bypasses global translational repression and underlies an increase in local translation through differential activity of eIF2B mediated by protein phosphatase 1. Ultrasensitive proteomics analysis of axons reveals 75 proteins translationally controlled via the Sema3A-p-eIF2α pathway. These include proteostasis- and actin cytoskeleton-related proteins but not canonical stress markers. Finally, we show that PERK signaling is needed for directional axon migration and visual pathway development in vivo. Thus, our findings reveal a noncanonical eIF2 signaling pathway that controls selective changes in axon translation and is required for neural wiring.


Asunto(s)
Factor 2B Eucariótico de Iniciación/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Neurogénesis , Células Ganglionares de la Retina/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Axones/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Factor 2B Eucariótico de Iniciación/genética , Femenino , Masculino , Neurogénesis/efectos de los fármacos , Fosforilación , Mapas de Interacción de Proteínas , Proteómica/métodos , Células Ganglionares de la Retina/efectos de los fármacos , Semaforina-3A/metabolismo , Semaforina-3A/farmacología , Transducción de Señal , Técnicas de Cultivo de Tejidos , Xenopus laevis/embriología , Xenopus laevis/metabolismo , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
4.
Brain ; 147(2): 649-664, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37703312

RESUMEN

The unfolded protein response (UPR) is rapidly gaining momentum as a therapeutic target for protein misfolding neurodegenerative diseases, in which its overactivation results in sustained translational repression leading to synapse loss and neurodegeneration. In mouse models of these disorders, from Alzheimer's to prion disease, modulation of the pathway-including by the licensed drug, trazodone-restores global protein synthesis rates with profound neuroprotective effects. However, the precise nature of the translational impairment, in particular the specific proteins affected in disease, and their response to therapeutic UPR modulation are poorly understood. We used non-canonical amino acid tagging (NCAT) to measure de novo protein synthesis in the brains of prion-diseased mice with and without trazodone treatment, in both whole hippocampus and cell-specifically. During disease the predominant nascent proteome changes occur in synaptic, cytoskeletal and mitochondrial proteins in both hippocampal neurons and astrocytes. Remarkably, trazodone treatment for just 2 weeks largely restored the whole disease nascent proteome in the hippocampus to that of healthy, uninfected mice, predominantly with recovery of proteins involved in synaptic and mitochondrial function. In parallel, trazodone treatment restored the disease-associated decline in synapses and mitochondria and their function to wild-type levels. In conclusion, this study increases our understanding of how translational repression contributes to neurodegeneration through synaptic and mitochondrial toxicity via depletion of key proteins essential for their function. Further, it provides new insights into the neuroprotective mechanisms of trazodone through reversal of this toxicity, relevant for the treatment of neurodegenerative diseases via translational modulation.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Enfermedades por Prión , Priones , Trazodona , Ratones , Animales , Priones/metabolismo , Proteoma/metabolismo , Proteoma/farmacología , Trazodona/farmacología , Trazodona/uso terapéutico , Trazodona/metabolismo , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Sinapsis/metabolismo , Enfermedad de Alzheimer/metabolismo
5.
Nucleic Acids Res ; 50(19): e112, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-35979952

RESUMEN

The assessment of transcriptome-wide ribosome binding to mRNAs is useful for studying the dynamic regulation of protein synthesis. Two methods frequently applied in eukaryotic cells that operate at different levels of resolution are polysome profiling, which reveals the distribution of ribosome loads across the transcriptome, and ribosome footprinting (also termed ribosome profiling or Ribo-Seq), which when combined with appropriate data on mRNA expression can reveal ribosome densities on individual transcripts. In this study we develop methods for relating the information content of these two methods to one another, by reconstructing theoretical polysome profiles from ribosome footprinting data. Our results validate both approaches as experimental tools. Although we show that both methods can yield highly consistent data, some published ribosome footprinting datasets give rise to reconstructed polysome profiles with non-physiological features. We trace these aberrant features to inconsistencies in RNA and Ribo-Seq data when compared to datasets yielding physiological polysome profiles, thereby demonstrating that modelled polysomes are useful for assessing global dataset properties such as its quality in a simple, visual approach. Aside from using polysome profile reconstructions on published datasets, we propose that this also provides a useful tool for validating new ribosome footprinting datasets in early stages of analyses.


Asunto(s)
Biosíntesis de Proteínas , Ribosomas , Ribosomas/genética , Ribosomas/metabolismo , Polirribosomas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcriptoma
6.
Artículo en Inglés | MEDLINE | ID: mdl-35921312

RESUMEN

The unfolded protein response has been increasingly implicated as an important pathological pathway and target for therapeutic intervention in neurodegeneration. The licensed antidepressant trazodone is one drug which has been proposed to act on this pathway and may therefore be a potential therapy. Previous examination of existing data for patients with dementia prescribed trazodone did not find a signal suggesting a disease modifying effect. Here we add to that literature by examining the electronic patient record of patients with dementia in Cambridgeshire UK. We found that trazodone is rarely prescribed and where it is used it is at a dose less than half that predicted to be disease modifying. We also found that patients prescribed trazodone had higher levels of neuropsychiatric symptoms and were relatively late in the disease course, likely beyond the optimal point for therapeutic intervention. We suggest it is therefore premature to discard potential therapies based on observational data alone, particularly when experimental medicine approaches to examine the effects of trazodone are feasible.


Asunto(s)
Demencia , Trazodona , Antidepresivos/uso terapéutico , Estudios de Cohortes , Demencia/tratamiento farmacológico , Humanos , Trazodona/farmacología , Trazodona/uso terapéutico
7.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35216136

RESUMEN

Dementia is a global medical and societal challenge; it has devastating personal, social and economic costs, which will increase rapidly as the world's population ages. Despite this, there are no disease-modifying treatments for dementia; current therapy modestly improves symptoms but does not change the outcome. Therefore, new treatments are urgently needed-particularly any that can slow down the disease's progression. Many of the neurodegenerative diseases that lead to dementia are characterised by common pathological responses to abnormal protein production and misfolding in brain cells, raising the possibility of the broad application of therapeutics that target these common processes. The unfolded protein response (UPR) is one such mechanism. The UPR is a highly conserved cellular stress response to abnormal protein folding and is widely dysregulated in neurodegenerative diseases. In this review, we describe the basic machinery of the UPR, as well as the evidence for its overactivation and pathogenicity in dementia, and for the marked neuroprotective effects of its therapeutic manipulation in murine models of these disorders. We discuss drugs identified as potential UPR-modifying therapeutic agents-in particular the licensed antidepressant trazodone-and we review epidemiological and trial data from their use in human populations. Finally, we explore future directions for investigating the potential benefit of using trazodone or similar UPR-modulating compounds for disease modification in patients with dementia.


Asunto(s)
Demencia/patología , Respuesta de Proteína Desplegada/fisiología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Demencia/tratamiento farmacológico , Humanos , Trazodona/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
8.
PLoS Biol ; 16(4): e2003611, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29630591

RESUMEN

Nitric oxide (NO) regulates neuronal function and thus is critical for tuning neuronal communication. Mechanisms by which NO modulates protein function and interaction include posttranslational modifications (PTMs) such as S-nitrosylation. Importantly, cross signaling between S-nitrosylation and prenylation can have major regulatory potential. However, the exact protein targets and resulting changes in function remain elusive. Here, we interrogated the role of NO-dependent PTMs and farnesylation in synaptic transmission. We found that NO compromises synaptic function at the Drosophila neuromuscular junction (NMJ) in a cGMP-independent manner. NO suppressed release and reduced the size of available vesicle pools, which was reversed by glutathione (GSH) and occluded by genetic up-regulation of GSH-generating and de-nitrosylating glutamate-cysteine-ligase and S-nitroso-glutathione reductase activities. Enhanced nitrergic activity led to S-nitrosylation of the fusion-clamp protein complexin (cpx) and altered its membrane association and interactions with active zone (AZ) and soluble N-ethyl-maleimide-sensitive fusion protein Attachment Protein Receptor (SNARE) proteins. Furthermore, genetic and pharmacological suppression of farnesylation and a nitrosylation mimetic mutant of cpx induced identical physiological and localization phenotypes as caused by NO. Together, our data provide evidence for a novel physiological nitrergic molecular switch involving S-nitrosylation, which reversibly suppresses farnesylation and thereby enhances the net-clamping function of cpx. These data illustrate a new mechanistic signaling pathway by which regulation of farnesylation can fine-tune synaptic release.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/metabolismo , Óxido Nítrico/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Adaptadoras del Transporte Vesicular/genética , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Encéfalo/metabolismo , GMP Cíclico/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Larva/genética , Larva/metabolismo , Proteínas del Tejido Nervioso/genética , Unión Neuromuscular/citología , Unión Neuromuscular/metabolismo , Fenotipo , Prenilación , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Transmisión Sináptica , Vesículas Sinápticas/metabolismo
9.
Nature ; 518(7538): 236-9, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25607368

RESUMEN

In the healthy adult brain synapses are continuously remodelled through a process of elimination and formation known as structural plasticity. Reduction in synapse number is a consistent early feature of neurodegenerative diseases, suggesting deficient compensatory mechanisms. Although much is known about toxic processes leading to synaptic dysfunction and loss in these disorders, how synaptic regeneration is affected is unknown. In hibernating mammals, cooling induces loss of synaptic contacts, which are reformed on rewarming, a form of structural plasticity. We have found that similar changes occur in artificially cooled laboratory rodents. Cooling and hibernation also induce a number of cold-shock proteins in the brain, including the RNA binding protein, RBM3 (ref. 6). The relationship of such proteins to structural plasticity is unknown. Here we show that synapse regeneration is impaired in mouse models of neurodegenerative disease, in association with the failure to induce RBM3. In both prion-infected and 5XFAD (Alzheimer-type) mice, the capacity to regenerate synapses after cooling declined in parallel with the loss of induction of RBM3. Enhanced expression of RBM3 in the hippocampus prevented this deficit and restored the capacity for synapse reassembly after cooling. RBM3 overexpression, achieved either by boosting endogenous levels through hypothermia before the loss of the RBM3 response or by lentiviral delivery, resulted in sustained synaptic protection in 5XFAD mice and throughout the course of prion disease, preventing behavioural deficits and neuronal loss and significantly prolonging survival. In contrast, knockdown of RBM3 exacerbated synapse loss in both models and accelerated disease and prevented the neuroprotective effects of cooling. Thus, deficient synapse regeneration, mediated at least in part by failure of the RBM3 stress response, contributes to synapse loss throughout the course of neurodegenerative disease. The data support enhancing cold-shock pathways as potential protective therapies in neurodegenerative disorders.


Asunto(s)
Frío , Respuesta al Choque por Frío/fisiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Plasticidad Neuronal , Fármacos Neuroprotectores , Proteínas de Unión al ARN/metabolismo , Sinapsis/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Proteínas y Péptidos de Choque por Frío/metabolismo , Modelos Animales de Enfermedad , Hibernación/fisiología , Hipocampo/metabolismo , Masculino , Ratones , Priones/fisiología , Proteínas de Unión al ARN/genética , Regeneración
10.
RNA ; 22(4): 623-35, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26857222

RESUMEN

The RNA exosome is essential for 3' processing of functional RNA species and degradation of aberrant RNAs in eukaryotic cells. Recent reports have defined the substrates of the exosome catalytic domains and solved the multimeric structure of the exosome complex. However, regulation of exosome activity remains poorly characterized, especially in response to physiological stress. Following the observation that cooling of mammalian cells results in a reduction in 40S:60S ribosomal subunit ratio, we uncover regulation of the nuclear exosome as a result of reduced temperature. Using human cells and an in vivo model system allowing whole-body cooling, we observe reduced EXOSC10 (hRrp6, Pm/Scl-100) expression in the cold. In parallel, both models of cooling increase global SUMOylation, leading to the identification of specific conjugation of SUMO1 to EXOSC10, a process that is increased by cooling. Furthermore, we define the major SUMOylation sites in EXOSC10 by mutagenesis and show that overexpression of SUMO1 alone is sufficient to suppress EXOSC10 abundance. Reducing EXOSC10 expression by RNAi in human cells correlates with the 3' preribosomal RNA processing defects seen in the cold as well as reducing the 40S:60S ratio, a previously uncharacterized consequence of EXOSC10 suppression. Together, this work illustrates that EXOSC10 can be modified by SUMOylation and identifies a physiological stress where this regulation is prevalent both in vitro and in vivo.


Asunto(s)
Exorribonucleasas/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Secuencia de Aminoácidos , Animales , Respuesta al Choque por Frío , Represión Enzimática , Exorribonucleasas/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Células HEK293 , Humanos , Ratones , Datos de Secuencia Molecular , Biosíntesis de Proteínas , ARN Ribosómico/metabolismo , Proteína SUMO-1/metabolismo , Sumoilación
11.
Brain ; 140(6): 1768-1783, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28430857

RESUMEN

See Mercado and Hetz (doi:10.1093/brain/awx107) for a scientific commentary on this article.Signalling through the PERK/eIF2α-P branch of the unfolded protein response plays a critical role in controlling protein synthesis rates in cells. This pathway is overactivated in brains of patients with Alzheimer’s disease and related disorders and has recently emerged as a promising therapeutic target for these currently untreatable conditions. Thus, in mouse models of neurodegenerative disease, prolonged overactivation of PERK/eIF2α-P signalling causes sustained attenuation of protein synthesis, leading to memory impairment and neuronal loss. Re-establishing translation rates by inhibition of eIF2α-P activity, genetically or pharmacologically, restores memory and prevents neurodegeneration and extends survival. However, the experimental compounds used preclinically are unsuitable for use in humans, due to associated toxicity or poor pharmacokinetic properties. To discover compounds that have anti-eIF2α-P activity suitable for clinical use, we performed phenotypic screens on a NINDS small molecule library of 1040 drugs. We identified two compounds, trazodone hydrochloride and dibenzoylmethane, which reversed eIF2α-P-mediated translational attenuation in vitro and in vivo. Both drugs were markedly neuroprotective in two mouse models of neurodegeneration, using clinically relevant doses over a prolonged period of time, without systemic toxicity. Thus, in prion-diseased mice, both trazodone and dibenzoylmethane treatment restored memory deficits, abrogated development of neurological signs, prevented neurodegeneration and significantly prolonged survival. In tauopathy-frontotemporal dementia mice, both drugs were neuroprotective, rescued memory deficits and reduced hippocampal atrophy. Further, trazodone reduced p-tau burden. These compounds therefore represent potential new disease-modifying treatments for dementia. Trazodone in particular, a licensed drug, should now be tested in clinical trials in patients.


Asunto(s)
Chalconas/farmacología , Demencia Frontotemporal/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Enfermedades por Prión/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Trazodona/farmacología , eIF-2 Quinasa/antagonistas & inhibidores , Animales , Conducta Animal , Chalconas/administración & dosificación , Modelos Animales de Enfermedad , Demencia Frontotemporal/complicaciones , Trastornos de la Memoria/etiología , Ratones , Fármacos Neuroprotectores/administración & dosificación , Enfermedades por Prión/complicaciones , Inhibidores de Proteínas Quinasas/administración & dosificación , Trazodona/administración & dosificación , Respuesta de Proteína Desplegada
12.
Nature ; 485(7399): 507-11, 2012 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-22622579

RESUMEN

The mechanisms leading to neuronal death in neurodegenerative disease are poorly understood. Many of these disorders, including Alzheimer's, Parkinson's and prion diseases, are associated with the accumulation of misfolded disease-specific proteins. The unfolded protein response is a protective cellular mechanism triggered by rising levels of misfolded proteins. One arm of this pathway results in the transient shutdown of protein translation, through phosphorylation of the α-subunit of eukaryotic translation initiation factor, eIF2. Activation of the unfolded protein response and/or increased eIF2α-P levels are seen in patients with Alzheimer's, Parkinson's and prion diseases, but how this links to neurodegeneration is unknown. Here we show that accumulation of prion protein during prion replication causes persistent translational repression of global protein synthesis by eIF2α-P, associated with synaptic failure and neuronal loss in prion-diseased mice. Further, we show that promoting translational recovery in hippocampi of prion-infected mice is neuroprotective. Overexpression of GADD34, a specific eIF2α-P phosphatase, as well as reduction of levels of prion protein by lentivirally mediated RNA interference, reduced eIF2α-P levels. As a result, both approaches restored vital translation rates during prion disease, rescuing synaptic deficits and neuronal loss, thereby significantly increasing survival. In contrast, salubrinal, an inhibitor of eIF2α-P dephosphorylation, increased eIF2α-P levels, exacerbating neurotoxicity and significantly reducing survival in prion-diseased mice. Given the prevalence of protein misfolding and activation of the unfolded protein response in several neurodegenerative diseases, our results suggest that manipulation of common pathways such as translational control, rather than disease-specific approaches, may lead to new therapies preventing synaptic failure and neuronal loss across the spectrum of these disorders.


Asunto(s)
Factor 2 Eucariótico de Iniciación/química , Factor 2 Eucariótico de Iniciación/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Fosfoproteínas/metabolismo , Priones/metabolismo , Biosíntesis de Proteínas , Proteínas Represoras/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Cinamatos/farmacología , Factor 2 Eucariótico de Iniciación/análisis , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores , Fosfoproteínas/análisis , Fosforilación , Proteínas PrPSc/análisis , Proteínas PrPSc/metabolismo , Proteínas PrPSc/toxicidad , Enfermedades por Prión/patología , Priones/biosíntesis , Priones/genética , Biosíntesis de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Proteínas Represoras/análisis , Proteínas Represoras/química , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología , Transmisión Sináptica/efectos de los fármacos , Tiourea/análogos & derivados , Tiourea/farmacología , Respuesta de Proteína Desplegada/fisiología
13.
J Neurochem ; 142(6): 812-826, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28643372

RESUMEN

Protein translation and folding are tightly controlled processes in all cells, by proteostasis, an important component of which is the unfolded protein response (UPR). During periods of endoplasmic reticulum stress because of protein misfolding, the UPR activates a coordinated response in which the PERK branch activation restricts translation, while a variety of genes involved with protein folding, degradation, chaperone expression and stress responses are induced through signaling of the other branches. Chronic overactivation of the UPR, particularly the PERK branch, is observed in the brains of patients in a number of protein misfolding neurodegenerative diseases, including Alzheimer's, and Parkinson's diseases and the tauopathies. Recently, numerous genetic and pharmacological studies in mice have demonstrated the effectiveness of inhibiting the UPR for eliciting therapeutic benefit and boosting memory. In particular, fine-tuning the level of PERK inhibition to provide neuroprotection without adverse side effects has emerged as a safe, effective approach. This includes the recent discovery of licensed drugs that can now be repurposed in clinical trials for new human treatments for dementia. This review provides an overview of the links between UPR overactivation and neurodegeneration in protein misfolding disorders. It discusses recent therapeutic approaches targeting this pathway, with a focus on treatments that fine-tune PERK signaling.

14.
Brain ; 139(Pt 8): 2113-21, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27190028

RESUMEN

Activation of the unfolded protein response is emerging as a common theme in protein-misfolding neurodegenerative diseases, with relevant markers observed in patient tissue and mouse models. Genetic and pharmacological manipulation of the pathway in several mouse models has shown that this is not a passive consequence of the neurodegeneration process. Rather, overactivation of the protein kinase RNA-like ER kinase (PERK, encoded by EIF2AK3) branch of the unfolded protein response directly contributes to disease pathogenesis through the critical reduction in neuronal protein synthesis rates, essential for learning and memory and for neuronal survival. The pharmacological inhibition of this process in these models is strikingly neuroprotective, resulting in the discovery of the first small molecule preventing neurodegeneration and clinical disease in vivo This now represents a potential generic approach for boosting memory and preventing neurodegeneration across the spectrum of these disorders, albeit with some exceptions, independent of disease-specific proteins. Targeting the unfolded protein response, and particularly PERK-branch mediated translational failure is thus an increasingly compelling strategy for new treatments for dementia and neurodegenerative disease.


Asunto(s)
Demencia/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos , eIF-2 Quinasa/metabolismo , Animales , Demencia/tratamiento farmacológico , Humanos , Ratones , Enfermedades Neurodegenerativas/tratamiento farmacológico
15.
Mol Cell Proteomics ; 13(10): 2584-92, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24973420

RESUMEN

We report a novel strategy for studying synaptic pathology by concurrently measuring levels of four SNARE complex proteins from individual brain tissue samples. This method combines affinity purification and mass spectrometry and can be applied directly for studies of SNARE complex proteins in multiple species or modified to target other key elements in neuronal function. We use the technique to demonstrate altered levels of presynaptic proteins in Alzheimer disease patients and prion-infected mice.


Asunto(s)
Envejecimiento/fisiología , Enfermedad de Alzheimer/fisiopatología , Espectrometría de Masas/métodos , Proteínas SNARE/metabolismo , Sinapsis/metabolismo , Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Cromatografía Liquida/métodos , Modelos Animales de Enfermedad , Humanos , Ratones , Enfermedades por Prión/metabolismo , Enfermedades por Prión/fisiopatología , Enfermedades por Prión/veterinaria , Proteómica/métodos , Sinapsis/patología
16.
J Biol Chem ; 289(29): 19862-8, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24860100

RESUMEN

Neurodegenerative diseases are characterized by the aggregation of misfolded proteins in the brain. Among these disorders are the prion diseases, which are transmissible, and in which the misfolded proteins ("prions") are also the infectious agent. Increasingly, it appears that misfolded proteins in Alzheimer and Parkinson diseases and the tauopathies also propagate in a "prion-like" manner. However, the association between prion formation, spread, and neurotoxicity is not clear. Recently, we showed that in prion disease, protein misfolding leads to neurodegeneration through dysregulation of generic proteostatic mechanisms, specifically, the unfolded protein response. Genetic and pharmacological manipulation of the unfolded protein response was neuroprotective despite continuing prion replication, hence dissociating this from neurotoxicity. The data have clear implications for treatment across the spectrum of these disorders, targeting pathogenic processes downstream of protein misfolding.


Asunto(s)
Enfermedades por Prión/etiología , Enfermedades por Prión/metabolismo , Priones/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Indoles/farmacología , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedades por Prión/tratamiento farmacológico , Priones/química , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología , Tauopatías/etiología , Tauopatías/metabolismo , Respuesta de Proteína Desplegada , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo
17.
Neuropathol Appl Neurobiol ; 41(4): 414-27, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25556298

RESUMEN

Recent evidence has placed the unfolded protein response (UPR) at the centre of pathological processes leading to neurodegenerative disease. The translational repression caused by UPR activation starves neurons of the essential proteins they need to function and survive. Restoration of protein synthesis, via genetic or pharmacological means, is neuroprotective in animal models, prolonging survival. This is of great interest due to the observation of UPR activation in the post mortem brains of patients with Alzheimer's, Parkinson's, tauopathies and prion diseases. Protein synthesis is also an essential step in the formation of new memories. Restoring translation in disease or increasing protein synthesis from basal levels has been shown to improve memory in numerous models. As neurodegenerative diseases often present with memory impairments, targeting the UPR to both provide neuroprotection and enhance memory provides an extremely exciting novel therapeutic target.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Respuesta de Proteína Desplegada , Animales , Estrés del Retículo Endoplásmico , Humanos , Memoria/fisiología
18.
Acta Neuropathol ; 130(5): 633-42, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26450683

RESUMEN

The PERK-eIF2α branch of the Unfolded Protein Response (UPR) mediates the transient shutdown of translation in response to rising levels of misfolded proteins in the endoplasmic reticulum. PERK and eIF2α activation are increasingly recognised in postmortem analyses of patients with neurodegenerative disorders, including Alzheimer's disease, the tauopathies and prion disorders. These are all characterised by the accumulation of misfolded disease-specific proteins in the brain in association with specific patterns of neuronal loss, but the role of UPR activation in their pathogenesis is unclear. In prion-diseased mice, overactivation of PERK-P/eIF2α-P signalling results in the sustained reduction in global protein synthesis, leading to synaptic failure, neuronal loss and clinical disease. Critically, restoring vital neuronal protein synthesis rates by inhibiting the PERK-eIF2α pathway, both genetically and pharmacologically, prevents prion neurodegeneration downstream of misfolded prion protein accumulation. Here we show that PERK-eIF2α-mediated translational failure is a key process leading to neuronal loss in a mouse model of frontotemporal dementia, where the misfolded protein is a form of mutant tau. rTg4510 mice, which overexpress the P301L tau mutation, show dysregulated PERK signalling and sustained repression of protein synthesis by 6 months of age, associated with onset of neurodegeneration. Treatment with the PERK inhibitor, GSK2606414, from this time point in mutant tau-expressing mice restores protein synthesis rates, protecting against further neuronal loss, reducing brain atrophy and abrogating the appearance of clinical signs. Further, we show that PERK-eIF2α activation also contributes to the pathological phosphorylation of tau in rTg4510 mice, and that levels of phospho-tau are lowered by PERK inhibitor treatment, providing a second mechanism of protection. The data support UPR-mediated translational failure as a generic pathogenic mechanism in protein-misfolding disorders, including tauopathies, that can be successfully targeted for prevention of neurodegeneration.


Asunto(s)
Adenina/análogos & derivados , Demencia Frontotemporal/tratamiento farmacológico , Indoles/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , eIF-2 Quinasa/antagonistas & inhibidores , Proteínas tau/metabolismo , Adenina/farmacología , Animales , Atrofia , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Demencia Frontotemporal/enzimología , Demencia Frontotemporal/patología , Humanos , Masculino , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Mutación , Neuronas/enzimología , Neuronas/patología , Tamaño de los Órganos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo , Proteínas tau/genética
20.
EMBO Mol Med ; 15(5): e17157, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36946385

RESUMEN

Neurodegenerative diseases are increasingly prevalent in the aging population, yet no disease-modifying treatments are currently available. Increasing the expression of the cold-shock protein RBM3 through therapeutic hypothermia is remarkably neuroprotective. However, systemic cooling poses a health risk, strongly limiting its clinical application. Selective upregulation of RBM3 at normothermia thus holds immense therapeutic potential. Here we identify a poison exon within the RBM3 gene that is solely responsible for its cold-induced expression. Genetic removal or antisense oligonucleotide (ASO)-mediated manipulation of this exon yields high RBM3 levels independent of cooling. Notably, a single administration of ASO to exclude the poison exon, using FDA-approved chemistry, results in long-lasting increased RBM3 expression in mouse brains. In prion-diseased mice, this treatment leads to remarkable neuroprotection, with prevention of neuronal loss and spongiosis despite high levels of disease-associated prion protein. Our promising results in mice support the possibility that RBM3-inducing ASOs might also deliver neuroprotection in humans in conditions ranging from acute brain injury to Alzheimer's disease.


Asunto(s)
Oligonucleótidos Antisentido , Venenos , Humanos , Ratones , Animales , Anciano , Temperatura , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacología , Proteínas de Unión al ARN/genética , Frío
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA