Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 13(1): 3236, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35688821

ABSTRACT

Dysregulated protein synthesis is a core pathogenic mechanism in Fragile X Syndrome (FX). The mGluR Theory of FX predicts that pathological synaptic changes arise from the excessive translation of mRNAs downstream of mGlu1/5 activation. Here, we use a combination of CA1 pyramidal neuron-specific TRAP-seq and proteomics to identify the overtranslating mRNAs supporting exaggerated mGlu1/5 -induced long-term synaptic depression (mGluR-LTD) in the FX mouse model (Fmr1-/y). Our results identify a significant increase in the translation of ribosomal proteins (RPs) upon mGlu1/5 stimulation that coincides with a reduced translation of long mRNAs encoding synaptic proteins. These changes are mimicked and occluded in Fmr1-/y neurons. Inhibiting RP translation significantly impairs mGluR-LTD and prevents the length-dependent shift in the translating population. Together, these results suggest that pathological changes in FX result from a length-dependent alteration in the translating population that is supported by excessive RP translation.


Subject(s)
Fragile X Syndrome , Receptors, Metabotropic Glutamate , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
2.
Sci Signal ; 13(644)2020 08 11.
Article in English | MEDLINE | ID: mdl-32788341

ABSTRACT

Chronic activation of the unfolded protein response (UPR), notably the branch comprising the kinase PERK and the translation initiation factor eIF2α, is a pathological feature of many neurodegenerative diseases caused by protein misfolding. Partial reduction of UPR signaling at the level of phosphorylated eIF2α is neuroprotective and avoids the pancreatic toxicity caused by full inhibition of PERK kinase activity. However, other stress pathways besides the UPR converge on phosphorylated eIF2α in the integrated stress response (ISR), which is critical to normal cellular function. We explored whether partial inhibition of PERK signaling may be a better therapeutic option. PERK-mediated phosphorylation of eIF2α requires its binding to the insert loop within PERK's kinase domain, which is, itself, phosphorylated at multiple sites. We found that, as expected, Akt mediates the phosphorylation of Thr799 in PERK. This phosphorylation event reduced eIF2α binding to PERK and selectively attenuated downstream signaling independently of PERK activity and the broader ISR. Induction of Thr799 phosphorylation with a small-molecule activator of Akt similarly reduced PERK signaling and increased both neuronal and animal survival without measurable pancreatic toxicity in a mouse model of prion disease. Thus, promoting PERK phosphorylation at Thr799 to partially down-regulate PERK-eIF2α signaling while avoiding widespread ISR inhibition may be a safe therapeutic approach in neurodegenerative disease.


Subject(s)
Disease Models, Animal , Eukaryotic Initiation Factor-2/metabolism , Prion Diseases/metabolism , Signal Transduction , eIF-2 Kinase/metabolism , Acetates/pharmacology , Animals , Benzopyrans/pharmacology , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , HEK293 Cells , Humans , Kaplan-Meier Estimate , Mice , Phosphorylation/drug effects , Prion Diseases/drug therapy , Unfolded Protein Response/drug effects
3.
Neuron ; 105(5): 855-866.e5, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31924446

ABSTRACT

Recent interest in astrocyte activation states has raised the fundamental question of how these cells, normally essential for synapse and neuronal maintenance, become pathogenic. Here, we show that activation of the unfolded protein response (UPR), specifically phosphorylated protein kinase R-like endoplasmic reticulum (ER) kinase (PERK-P) signaling-a pathway that is widely dysregulated in neurodegenerative diseases-generates a distinct reactivity state in astrocytes that alters the astrocytic secretome, leading to loss of synaptogenic function in vitro. Further, we establish that the same PERK-P-dependent astrocyte reactivity state is harmful to neurons in vivo in mice with prion neurodegeneration. Critically, targeting this signaling exclusively in astrocytes during prion disease is alone sufficient to prevent neuronal loss and significantly prolongs survival. Thus, the astrocyte reactivity state resulting from UPR over-activation is a distinct pathogenic mechanism that can by itself be effectively targeted for neuroprotection.


Subject(s)
Astrocytes/metabolism , Eukaryotic Initiation Factor-2B/metabolism , Neurodegenerative Diseases/metabolism , Prion Diseases/metabolism , Synapses/metabolism , Unfolded Protein Response/physiology , eIF-2 Kinase/metabolism , Animals , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Memory , Mice , Phosphorylation , Protein Biosynthesis , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Signal Transduction , Thapsigargin/pharmacology , Transcriptome , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects
4.
Brain ; 140(6): 1768-1783, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28430857

ABSTRACT

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.


Subject(s)
Chalcones/pharmacology , Frontotemporal Dementia/drug therapy , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Prion Diseases/drug therapy , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Trazodone/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Animals , Behavior, Animal , Chalcones/administration & dosage , Disease Models, Animal , Frontotemporal Dementia/complications , Memory Disorders/etiology , Mice , Neuroprotective Agents/administration & dosage , Prion Diseases/complications , Protein Kinase Inhibitors/administration & dosage , Trazodone/administration & dosage , Unfolded Protein Response
5.
Biochem J ; 433(1): 19-29, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21158739

ABSTRACT

One of the major current challenges to both medicine and neuroscience is the treatment of neurodegenerative diseases, which pose an ever-increasing medical, social and economic burden in the developed world. These disorders, which include Alzheimer's, Huntington's and Parkinson's diseases, and the rarer prion diseases, are separate entities clinically but have common features, including aggregates of misfolded proteins and varying patterns of neurodegeneration. A key barrier to effective treatment is that patients present clinically with advanced, irreversible, neuronal loss. Critically, mechanisms of neurotoxicity are poorly understood. Prevention of neuronal loss, ideally by targeting underlying pathogenic mechanisms, must be the aim of therapy. The present review describes the rationale and experimental approaches that have allowed such prevention, rescuing neurons in mice with prion disease. This rescue cured animals of a rapidly fatal neurodegenerative condition, resulting in symptom-free survival for their natural lifespan. Early pathological changes were reversed; behavioural, cognitive and neurophysiological deficits were recovered; and there was no neuronal loss. This was achieved by targeting the central pathogenic process in prion disease rather than the presumed toxic species, first by proof-of-principle experiments in transgenic mice and then by treatment using RNA interference for gene knockdown. The results have been a new therapeutic target for prion disease, further insight into mechanisms of prion neurotoxicity and the discovery of a window of reversibility in neuronal damage. Furthermore, the work gives rise to new concepts for treatment strategies for other neurodegenerative disorders, and highlights the need for clinical detection of early neuronal dysfunction, so that similar early rescue can also be achieved for these disorders.


Subject(s)
Neurons/pathology , Prion Diseases/therapy , RNA Interference , Animals , Disease-Free Survival , Mice , Neuroprotective Agents , Prion Diseases/prevention & control , Remission Induction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...