phosphorylated eif2α
Recently Published Documents


TOTAL DOCUMENTS

18
(FIVE YEARS 6)

H-INDEX

9
(FIVE YEARS 1)

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Eva Ramos-Fernández ◽  
Macarena S. Arrázola ◽  
Carolina A. Oliva ◽  
Sebastián B. Arredondo ◽  
Lorena Varela-Nallar ◽  
...  

AbstractWnt signaling plays a key role in neurodevelopment and neuronal maturation. Specifically, Wnt5a stimulates postsynaptic assemblies, increases glutamatergic neurotransmission and, through calcium signaling, generates nitric oxide (NO). Trying to unveil the molecular pathway triggering these postsynaptic effects, we found that Wnt5a treatment induces a time-dependent increases in the length of the postsynaptic density (PSD), elicits novel synaptic contacts and facilitates F-actin flow both in in vitro and ex vivo models. These effects were partially abolished by the inhibition of the Heme-regulated eukaryotic initiation factor 2α (HRI) kinase, a kinase which phosphorylates the initiation translational factor eIF2α. When phosphorylated, eIF2α normally avoids the translation of proteins not needed during stress conditions, in order to avoid unnecessary energetic expenses. However, phosphorylated eIF2α promotes the translation of some proteins with more than one open reading frame in its 5′ untranslated region. One of these proteins targeted by Wnt-HRI-eIF2α mediated translation is the GluN2B subunit of the NMDA receptor. The identified increase in GluN2B expression correlated with increased NMDA receptor function. Considering that NMDA receptors are crucial for excitatory synaptic transmission, the molecular pathway described here contributes to the understanding of the fast and plastic translational mechanisms activated during learning and memory processes.


2021 ◽  
Vol 14 (668) ◽  
pp. eabc5429
Author(s):  
Mauricio M. Oliveira ◽  
Mychael V. Lourenco ◽  
Francesco Longo ◽  
Nicole P. Kasica ◽  
Wenzhong Yang ◽  
...  

Neuronal protein synthesis is essential for long-term memory consolidation, and its dysregulation is implicated in various neurodegenerative disorders, including Alzheimer’s disease (AD). Cellular stress triggers the activation of protein kinases that converge on the phosphorylation of eukaryotic translation initiation factor 2α (eIF2α), which attenuates mRNA translation. This translational inhibition is one aspect of the integrated stress response (ISR). We found that postmortem brain tissue from AD patients showed increased phosphorylation of eIF2α and reduced abundance of eIF2B, another key component of the translation initiation complex. Systemic administration of the small-molecule compound ISRIB (which blocks the ISR downstream of phosphorylated eIF2α) rescued protein synthesis in the hippocampus, measures of synaptic plasticity, and performance on memory-associated behavior tests in wild-type mice cotreated with salubrinal (which inhibits translation by inducing eIF2α phosphorylation) and in both β-amyloid-treated and transgenic AD model mice. Thus, attenuating the ISR downstream of phosphorylated eIF2α may restore hippocampal protein synthesis and delay cognitive decline in AD patients.


2020 ◽  
Author(s):  
Karin Eytan ◽  
Ziv Versano ◽  
Moshe Leitner ◽  
Shoshana Paglin ◽  
Amos Toren ◽  
...  

AbstractCurrent therapies offer only a short relief for patients with pediatric glioblastoma (PED-GBM). Therefore, expanding treatment options for this fatal disease is of utmost importance. We found that PED-GBM cell lines, originated from diffuse intrinsic pontine glioma expressing H3K27M mutation (DIPG), or from hemispheric glioma expressing H3G34R mutation, are sensitive to combinations of histone deacetylase and PARP-1 inhibitors (vorinostat with either olaparib or veliparib). These combinations led to an enhanced decrease in their survival, and to increased phosphorylation of eIF2α. Experiments with the S51D phosphomimetic variant of eIF2α and with brain-penetrating inhibitors of phosphorylated eIF2α (p-eIF2α) dephospohrylation, salubrinal and raphin1, showed that increased eIF2α phosphorylation diminished PED-GBM cell survival and sensitized them to PARP-1 inhibitors as well as to ionizing irradiation, which is the main treatment modality in these patients. PED-GBM cells were also remarkably more sensitive to combination of vorinostat and PARP-1 inhibitors and to salubrinal and raphin1 than normal human astrocytes and fibroblasts.Importantly, although the overall effect of increased eIF2α phosphorylation was a reduced survival of PED-GBM cells, it also increased the cellular level of MTH1, an enzyme that protects treated cells against the incorporation of oxidized nucleotides into nucleic acids, resulting in an enhanced decrease in cell survival in response to the combination of salubrinal and MTH1 inhibitor, TH588.Our results indicate that combinations of the FDA approved drugs, vorinostat and either veliparib or olaparib, could potentially be included in PED-GBM treatment protocols and that the effect of salubrinal and raphin1 on PED-GBM survival warrants further evaluation.


2020 ◽  
Vol 13 (644) ◽  
pp. eabb4749
Author(s):  
Daniel T. Hughes ◽  
Mark Halliday ◽  
Heather L. Smith ◽  
Nicholas C. Verity ◽  
Colin Molloy ◽  
...  

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.


2020 ◽  
Vol 295 (19) ◽  
pp. 6344-6356 ◽  
Author(s):  
Aastha Kapoor ◽  
Carolyn G. Chen ◽  
Renato V. Iozzo

Endorepellin, the C-terminal fragment of the heparan sulfate proteoglycan perlecan, influences various signaling pathways in endothelial cells by binding to VEGFR2. In this study, we discovered that soluble endorepellin activates the canonical stress signaling pathway consisting of PERK, eIF2α, ATF4, and GADD45α. Specifically, endorepellin evoked transient activation of VEGFR2, which, in turn, phosphorylated PERK at Thr980. Subsequently, PERK phosphorylated eIF2α at Ser51, upregulating its downstream effector proteins ATF4 and GADD45α. RNAi-mediated knockdown of PERK or eIF2α abrogated the endorepellin-mediated up-regulation of GADD45α, the ultimate effector protein of this stress signaling cascade. To functionally validate these findings, we utilized an ex vivo model of angiogenesis. Exposure of the aortic rings embedded in 3D fibrillar collagen to recombinant endorepellin for 2–4 h activated PERK and induced GADD45α vis à vis vehicle-treated counterparts. Similar effects were obtained with the established cellular stress inducer tunicamycin. Notably, chronic exposure of aortic rings to endorepellin for 7–9 days markedly suppressed vessel sprouting, an angiostatic effect that was rescued by blocking PERK kinase activity. Our findings unravel a mechanism by which an extracellular matrix protein evokes stress signaling in endothelial cells, which leads to angiostasis.


2019 ◽  
Vol 93 (9) ◽  
Author(s):  
Audra J. Charron ◽  
Stephen L. Ward ◽  
Brian J. North ◽  
Stacey Ceron ◽  
David A. Leib

ABSTRACTHerpes simplex virus 1 (HSV-1) cycles between phases of latency in sensory neurons and replication in mucosal sites. HSV-1 encodes two key proteins that antagonize the shutdown of host translation, US11 through preventing PKR activation and ICP34.5 through mediating dephosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α). While profound attenuation of ICP34.5 deletion mutants has been repeatedly demonstrated, a role for US11 in HSV-1 pathogenesis remains unclear. We therefore generated an HSV-1 strain 17 US11-null virus and examined its propertiesin vitroandin vivo. In U373 glioblastoma cells, US11 cooperated with ICP34.5 to prevent eIF2α phosphorylation late in infection. However, the effect was muted in human corneal epithelial cells (HCLEs), which did not accumulate phosphorylated eIF2α unless both US11 and ICP34.5 were absent. Low levels of phosphorylated eIF2α correlated with continued protein synthesis and with the ability of virus lacking US11 to overcome antiviral immunity in HCLE and U373 cells. Neurovirulence following intracerebral inoculation of mice was not affected by the deletion of US11. In contrast, the time to endpoint criteria following corneal infection was greater for the US11-null virus than for the wild-type virus. Replication in trigeminal ganglia and periocular tissue was promoted by US11, as was periocular disease. The establishment of latency and the frequency of virus reactivation from trigeminal ganglia were unaffected by US11 deletion, although emergence of the US11-null virus occurred with slowed kinetics. Considered together, the data indicate that US11 facilitates the countering of antiviral response of infected cells and promotes the efficient emergence of virus following reactivation.IMPORTANCEAlphaherpesviruses are ubiquitous DNA viruses and include the human pathogens herpes simplex virus 1 (HSV-1) and HSV-2 and are significant causes of ulcerative mucosal sores, infectious blindness, encephalitis, and devastating neonatal disease. Successful primary infection and persistent coexistence with host immune defenses are dependent on the ability of these viruses to counter the antiviral response. HSV-1 and HSV-2 and other primate viruses within theSimplexvirusgenus encode US11, an immune antagonist that promotes virus production by preventing shutdown of protein translation. Here we investigated the impact of US11 deletion on HSV-1 growthin vitroand pathogenesisin vivo. This work supports a role for US11 in pathogenesis and emergence from latency, elucidating immunomodulation by this medically important cohort of viruses.


Yeast ◽  
2017 ◽  
Vol 34 (9) ◽  
pp. 371-382 ◽  
Author(s):  
Su Jung Lee ◽  
Rashmi Ramesh ◽  
Valerie de Boor ◽  
Jan M. Gebler ◽  
Richard C. Silva ◽  
...  

2010 ◽  
Vol 30 (8) ◽  
pp. 2006-2016 ◽  
Author(s):  
Karen A. Wehner ◽  
Sylvia Schütz ◽  
Peter Sarnow

ABSTRACT Cells possess mechanisms that permit survival and recovery from stress, several of which regulate the phosphorylation of eukaryotic translation initiation factor 2α (eIF2α). We identified the human OGFOD1 protein as a novel stress granule component that regulates the phosphorylation of eIF2α and the resumption of translation in cells recovering from arsenite-induced stress. Coimmunoprecipitation studies revealed that OGFOD1 associates with a small subset of stress granule proteins (G3BP1, USP10, Caprin1, and YB-1) and the ribosome in both unstressed and stressed cells. Overexpression of OGFOD1 led to increased abundance of phosphorylated eIF2α, both in unstressed cells and in cells exposed to arsenite-induced stress, and to accelerated apoptosis during stress. Conversely, knockdown of OGFOD1 resulted in smaller amounts of phosphorylated eIF2α and a faster accumulation of polyribosomes in cells recovering from stress. Finally, OGFOD1 interacted with both eIF2α and the eIF2α kinase heme-regulated inhibitor (HRI), which was identified as a novel stress granule resident. These findings argue that OGFOD1 plays important proapoptotic roles in the regulation of translation and HRI-mediated phosphorylation of eIF2α in cells subjected to arsenite-induced stress.


Sign in / Sign up

Export Citation Format

Share Document