Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nature ; 518(7538): 236-9, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25607368

RESUMO

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.


Assuntos
Temperatura Baixa , Resposta ao Choque Frio/fisiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Plasticidade Neuronal , Fármacos Neuroprotetores , Proteínas de Ligação a RNA/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/metabolismo , Animais , Proteínas e Peptídeos de Choque Frio/metabolismo , Modelos Animais de Doenças , Hibernação/fisiologia , Hipocampo/metabolismo , Masculino , Camundongos , Príons/fisiologia , Proteínas de Ligação a RNA/genética , Regeneração
2.
Brain ; 140(6): 1768-1783, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28430857

RESUMO

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.


Assuntos
Chalconas/farmacologia , Demência Frontotemporal/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Doenças Priônicas/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Trazodona/farmacologia , eIF-2 Quinase/antagonistas & inibidores , Animais , Comportamento Animal , Chalconas/administração & dosagem , Modelos Animais de Doenças , Demência Frontotemporal/complicações , Transtornos da Memória/etiologia , Camundongos , Fármacos Neuroprotetores/administração & dosagem , Doenças Priônicas/complicações , Inibidores de Proteínas Quinases/administração & dosagem , Trazodona/administração & dosagem , Resposta a Proteínas não Dobradas
3.
Nature ; 485(7399): 507-11, 2012 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-22622579

RESUMO

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.


Assuntos
Fator de Iniciação 2 em Eucariotos/química , Fator de Iniciação 2 em Eucariotos/metabolismo , Doenças Neurodegenerativas/metabolismo , Fosfoproteínas/metabolismo , Príons/metabolismo , Biossíntese de Proteínas , Proteínas Repressoras/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Cinamatos/farmacologia , Fator de Iniciação 2 em Eucariotos/análise , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/patologia , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores , Fosfoproteínas/análise , Fosforilação , Proteínas PrPSc/análise , Proteínas PrPSc/metabolismo , Proteínas PrPSc/toxicidade , Doenças Priônicas/patologia , Príons/biossíntese , Príons/genética , Biossíntese de Proteínas/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo , Proteínas Repressoras/análise , Proteínas Repressoras/química , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia , Transmissão Sináptica/efeitos dos fármacos , Tioureia/análogos & derivados , Tioureia/farmacologia , Resposta a Proteínas não Dobradas/fisiologia
4.
J Biol Chem ; 289(29): 19862-8, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24860100

RESUMO

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.


Assuntos
Doenças Priônicas/etiologia , Doenças Priônicas/metabolismo , Príons/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Indóis/farmacologia , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Doenças Priônicas/tratamento farmacológico , Príons/química , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Tauopatias/etiologia , Tauopatias/metabolismo , Resposta a Proteínas não Dobradas , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo
5.
Acta Neuropathol ; 130(5): 633-42, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26450683

RESUMO

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.


Assuntos
Adenina/análogos & derivados , Demência Frontotemporal/tratamento farmacológico , Indóis/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , eIF-2 Quinase/antagonistas & inibidores , Proteínas tau/metabolismo , Adenina/farmacologia , Animais , Atrofia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Demência Frontotemporal/enzimologia , Demência Frontotemporal/patologia , Humanos , Masculino , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Mutação , Neurônios/enzimologia , Neurônios/patologia , Tamanho do Órgão , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , eIF-2 Quinase/metabolismo , Proteínas tau/genética
6.
Curr Issues Mol Biol ; 12(2): 119-27, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19767655

RESUMO

There are two central phenomena in prion disease: prion replication and prion neurotoxicity. Underlying them both is the conversion of a host-encoded ubiquitously expressed protein, prion protein (PrP(C)), into a partially-protease resistant isoform, PrP(Sc), which accumulates in the brain. PrP(Sc) is associated with both pathology and infectivity. In the absence of PrP(C), PrP(Sc) cannot be generated and PrP-null mice do not propagate infectivity or develop pathology on infection with scrapie. However, while PrP(C) expression is fundamental to both prion infectivity and neurodegeneration, the uncoupling of these processes is a growing concept in the field. This dissociation is evident in subclinical states of prion infection, where PrP(Sc) levels are high in the absence of disease, and in transgenic mice experiments, where, despite extra-neuronal PrP(Sc) accumulation, even in very high amounts, there is no neurotoxicity. Both these models have further implications. Thus depleting PrP(C) from neurons (but not glia) of prion-infected mice prevents clinical disease, and detaching it from the surface of cells by removing its anchor does the same. The uncoupling toxicity and infectivity has implications for the nature of the neurotoxic species; these mouse models suggest that the site for the generation of this species is intra-neuronal. This review considers the role of neuronal surface-expressed PrP(C) in mediating toxicity in prion infection, and the dissociation of this from the deposition of PrP(Sc).


Assuntos
Glicosilfosfatidilinositóis/metabolismo , Proteínas PrPC/metabolismo , Scrapie/metabolismo , Animais , Glicosilfosfatidilinositóis/genética , Humanos , Proteínas PrPC/genética , Doenças Priônicas/genética , Doenças Priônicas/metabolismo , Scrapie/genética
7.
Sci Signal ; 13(644)2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32788341

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Fator de Iniciação 2 em Eucariotos/metabolismo , Doenças Priônicas/metabolismo , Transdução de Sinais , eIF-2 Quinase/metabolismo , Acetatos/farmacologia , Animais , Benzopiranos/farmacologia , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Células HEK293 , Humanos , Estimativa de Kaplan-Meier , Camundongos , Fosforilação/efeitos dos fármacos , Doenças Priônicas/tratamento farmacológico , Resposta a Proteínas não Dobradas/efeitos dos fármacos
8.
Biochim Biophys Acta ; 1779(4): 217-29, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18316045

RESUMO

Elongation of the poly(A) tails of specific mRNAs in the cytoplasm is a crucial regulatory step in oogenesis and early development of many animal species. The best studied example is the regulation of translation by cytoplasmic polyadenylation elements (CPEs) in the 3' untranslated region of mRNAs involved in Xenopus oocyte maturation. In this review we discuss the mechanism of translational control by the CPE binding protein (CPEB) in Xenopus oocytes as follows: 1. The cytoplasmic polyadenylation machinery such as CPEB, the subunits of cleavage and polyadenylation specificity factor (CPSF), symplekin, Gld-2 and poly(A) polymerase (PAP). 2. The signal transduction that leads to the activation of CPE-mediated polyadenylation during oocyte maturation, including the potential roles of kinases such as MAPK, Aurora A, CamKII, cdk1/Ringo and cdk1/cyclin B. 3. The role of deadenylation and translational repression, including the potential involvement of PARN, CCR4/NOT, maskin, pumilio, Xp54 (Ddx6, Rck), other P-body components and isoforms of the cap binding initiation factor eIF4E. Finally we discuss some of the remaining questions regarding the mechanisms of translational regulation by cytoplasmic polyadenylation and give our view on where our knowledge is likely to be expanded in the near future.


Assuntos
Citoplasma/metabolismo , Oócitos/metabolismo , Poli A/metabolismo , Poliadenilação/fisiologia , Polinucleotídeo Adenililtransferase/metabolismo , Biossíntese de Proteínas/fisiologia , Proteínas de Ligação a RNA/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Feminino , Oócitos/citologia , Oogênese/fisiologia , Proteínas Quinases/metabolismo , Transdução de Sinais/fisiologia , Xenopus
9.
Sci Transl Med ; 5(206): 206ra138, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-24107777

RESUMO

During prion disease, an increase in misfolded prion protein (PrP) generated by prion replication leads to sustained overactivation of the branch of the unfolded protein response (UPR) that controls the initiation of protein synthesis. This results in persistent repression of translation, resulting in the loss of critical proteins that leads to synaptic failure and neuronal death. We have previously reported that localized genetic manipulation of this pathway rescues shutdown of translation and prevents neurodegeneration in a mouse model of prion disease, suggesting that pharmacological inhibition of this pathway might be of therapeutic benefit. We show that oral treatment with a specific inhibitor of the kinase PERK (protein kinase RNA-like endoplasmic reticulum kinase), a key mediator of this UPR pathway, prevented UPR-mediated translational repression and abrogated development of clinical prion disease in mice, with neuroprotection observed throughout the mouse brain. This was the case for animals treated both at the preclinical stage and also later in disease when behavioral signs had emerged. Critically, the compound acts downstream and independently of the primary pathogenic process of prion replication and is effective despite continuing accumulation of misfolded PrP. These data suggest that PERK, and other members of this pathway, may be new therapeutic targets for developing drugs against prion disease or other neurodegenerative diseases where the UPR has been implicated.


Assuntos
Adenina/análogos & derivados , Indóis/administração & dosagem , Indóis/farmacologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/prevenção & controle , Doenças Priônicas/tratamento farmacológico , Doenças Priônicas/prevenção & controle , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Adenina/administração & dosagem , Adenina/sangue , Adenina/farmacologia , Adenina/uso terapêutico , Administração Oral , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Indóis/sangue , Indóis/uso terapêutico , Camundongos , Degeneração Neural/enzimologia , Degeneração Neural/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fosforilação/efeitos dos fármacos , Doenças Priônicas/enzimologia , Doenças Priônicas/patologia , Príons , Biossíntese de Proteínas/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/metabolismo
10.
Biol Cell ; 99(5): 239-50, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17241108

RESUMO

BACKGROUND INFORMATION: Maskin is a member of the TACC (transforming acidic coiled-coil) domain proteins found in Xenopus laevis oocytes and embryos. It has been implicated in the co-ordination of the spindle and has been reported to mediate translational repression of cyclin B1 mRNA. RESULTS: In the present study, we report that maskin mRNA is translationally repressed at the level of initiation in stage 4 oocytes and becomes activated in stage 6 oocytes. The translational repression of maskin mRNA correlates with the presence of a short poly(A) tail on this mRNA in stage 4 oocytes. The 3'-UTR (untranslated region) of maskin can confer the translational regulation to a reporter mRNA, and so can the 3'-UTR of human TACC3. A conserved GUCU repeat element was found to repress translation in both stage 4 and stage 6 oocytes, but deletion of this element did not abrogate repression in stage 4 oocytes. UV cross-linking experiments indicated that overlapping sets of proteins bind efficiently to both the maskin and the cyclin B1 3'-UTRs. As reported previously, CPEB [CPE (cytoplasmic polyadenylation element)-binding protein] binds to the cyclin B1 3'-UTR, but its binding to the maskin 3'-UTR is minimal. By RNA affinity chromatography and MS, we identified the EDEN-BP [EDEN (embryonic deadenylation element)-binding protein] as one of the proteins binding to both the maskin and the cyclin B1 3'-UTRs. CONCLUSIONS: Maskin mRNA is translationally regulated by at least two repressor elements and an activation element. One of the repessor elements is the evolutionarily conserved GUCU repeat. EDEN-BP binds to both the maskin and cyclin B1 3'-UTRs, indicating it may be involved in the deadenylation of these mRNAs.


Assuntos
Regiões 3' não Traduzidas/genética , Regulação da Expressão Gênica , Biossíntese de Proteínas/genética , Fatores de Transcrição/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Ciclina B/metabolismo , Ciclina B1 , Dados de Sequência Molecular , Oócitos/metabolismo , Oogênese , Poli A/metabolismo , Polirribossomos/metabolismo , Ligação Proteica , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/química , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA