Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Translational control of mGluR-dependent long-term depression and object-place learning by eIF2α

Abstract

At hippocampal synapses, activation of group I metabotropic glutamate receptors (mGluRs) induces long-term depression (LTD), which requires new protein synthesis. However, the underlying mechanism remains elusive. Here we describe the translational program that underlies mGluR-LTD and identify the translation factor eIF2α as its master effector. Genetically reducing eIF2α phosphorylation, or specifically blocking the translation controlled by eIF2α phosphorylation, prevented mGluR-LTD and the internalization of surface AMPA receptors (AMPARs). Conversely, direct phosphorylation of eIF2α, bypassing mGluR activation, triggered a sustained LTD and removal of surface AMPARs. Combining polysome profiling and RNA sequencing, we identified the mRNAs translationally upregulated during mGluR-LTD. Translation of one of these mRNAs, oligophrenin-1, mediates the LTD induced by eIF2α phosphorylation. Mice deficient in phospho-eIF2α–mediated translation are impaired in object-place learning, a behavioral task that induces hippocampal mGluR-LTD in vivo. Our findings identify a new model of mGluR-LTD, which promises to be of value in the treatment of mGluR-LTD-linked cognitive disorders.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Deficient eIF2α phosphorylation selectively prevents protein synthesis-dependent mGluR-LTD.
Figure 2: Direct stimulation of eIF2α phosphorylation induces LTD.
Figure 3: mGluR-LTD requires eIF2α-mediated translational control.
Figure 4: Activation of mGluR selectively promotes translation of Ophn1 mRNA by eIF2α phosphorylation.
Figure 5: eIF2α phosphorylation selectively mediates the translation of OPHN1 required for mGluR-LTD.
Figure 6: At hippocampal synapses phosphorylation of eIF2α and surface GluR1 immunoreactivity are negatively correlated.
Figure 7: Increased eIF2α-mediated translational control is needed for successful learning of novel object-space configuration.

Similar content being viewed by others

References

  1. Malenka, R.C. & Bear, M.F. LTP and LTD: an embarrassment of riches. Neuron 44, 5–21 (2004).

    Article  CAS  Google Scholar 

  2. Neves, G., Cooke, S.F. & Bliss, T.V. Synaptic plasticity, memory and the hippocampus: a neural network approach to causality. Nat. Rev. Neurosci. 9, 65–75 (2008).

    Article  CAS  Google Scholar 

  3. Luscher, C. & Huber, K.M. Group 1 mGluR-dependent synaptic long-term depression: mechanisms and implications for circuitry and disease. Neuron 65, 445–459 (2010).

    Article  CAS  Google Scholar 

  4. Collingridge, G.L., Peineau, S., Howland, J.G. & Wang, Y.T. Long-term depression in the CNS. Nat. Rev. Neurosci. 11, 459–473 (2010).

    Article  CAS  Google Scholar 

  5. Gladding, C.M., Fitzjohn, S.M. & Molnar, E. Metabotropic glutamate receptor-mediated long-term depression: molecular mechanisms. Pharmacol. Rev. 61, 395–412 (2009).

    Article  CAS  Google Scholar 

  6. Huber, K.M., Kayser, M.S. & Bear, M.F. Role for rapid dendritic protein synthesis in hippocampal mGluR-dependent long-term depression. Science 288, 1254–1257 (2000).

    Article  CAS  Google Scholar 

  7. Costa-Mattioli, M., Sossin, W.S., Klann, E. & Sonenberg, N. Translational Control of Long-Lasting Synaptic Plasticity and Memory. Neuron 61, 10–26 (2009).

    Article  CAS  Google Scholar 

  8. Sonenberg, N. & Hinnebusch, A.G. Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell 136, 731–745 (2009).

    Article  CAS  Google Scholar 

  9. Laplante, M. & Sabatini, D.M. mTOR signaling in growth control and disease. Cell 149, 274–293 (2012).

    Article  CAS  Google Scholar 

  10. Hou, L. & Klann, E. Activation of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin signaling pathway is required for metabotropic glutamate receptor-dependent long-term depression. J. Neurosci. 24, 6352–6361 (2004).

    Article  CAS  Google Scholar 

  11. Osterweil, E.K., Krueger, D.D., Reinhold, K. & Bear, M.F. Hypersensitivity to mGluR5 and ERK1/2 leads to excessive protein synthesis in the hippocampus of a mouse model of fragile X syndrome. J. Neurosci. 30, 15616–15627 (2010).

    Article  CAS  Google Scholar 

  12. Auerbach, B.D., Osterweil, E.K. & Bear, M.F. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature 480, 63–68 (2011).

    Article  CAS  Google Scholar 

  13. Dever, T.E. Gene-specific regulation by general translation factors. Cell 108, 545–556 (2002).

    Article  CAS  Google Scholar 

  14. Hinnebusch, A.G. Translational regulation of GCN4 and the general amino acid control of yeast. Annu. Rev. Microbiol. 59, 407–450 (2005).

    Article  CAS  Google Scholar 

  15. Goh, J.J. & Manahan-Vaughan, D. Spatial object recognition enables endogenous LTD that curtails LTP in the mouse hippocampus. Cereb. Cortex 23, 1118–1125 (2013).

    Article  Google Scholar 

  16. Goh, J.J. & Manahan-Vaughan, D. Endogenous hippocampal LTD that is enabled by spatial object recognition requires activation of NMDA receptors and the metabotropic glutamate receptor, mGlu5. Hippocampus 23, 129–138 (2013).

    Article  CAS  Google Scholar 

  17. Scheuner, D. et al. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol. Cell 7, 1165–1176 (2001).

    Article  CAS  Google Scholar 

  18. Bateup, H.S., Takasaki, K.T., Saulnier, J.L., Denefrio, C.L. & Sabatini, B.L. Loss of Tsc1 in vivo impairs hippocampal mGluR-LTD and increases excitatory synaptic function. J. Neurosci. 31, 8862–8869 (2011).

    Article  CAS  Google Scholar 

  19. Huber, K.M., Roder, J.C. & Bear, M.F. Chemical induction of mGluR5- and protein synthesis–dependent long-term depression in hippocampal area CA1. J. Neurophysiol. 86, 321–325 (2001).

    Article  CAS  Google Scholar 

  20. Morishita, W., Marie, H. & Malenka, R.C. Distinct triggering and expression mechanisms underlie LTD of AMPA and NMDA synaptic responses. Nat. Neurosci. 8, 1043–1050 (2005).

    Article  CAS  Google Scholar 

  21. Snyder, E.M. et al. Internalization of ionotropic glutamate receptors in response to mGluR activation. Nat. Neurosci. 4, 1079–1085 (2001).

    Article  CAS  Google Scholar 

  22. Boyce, M. et al. A selective inhibitor of eIF2alpha dephosphorylation protects cells from ER stress. Science 307, 935–939 (2005).

    Article  CAS  Google Scholar 

  23. Robert, F. et al. Initiation of Protein Synthesis by Hepatitis C Virus Is Refractory to Reduced eIF2·GTP·Met-tRNAiMet Ternary Complex Availability. Mol. Biol. Cell 17, 4632–4644 (2006).

    Article  CAS  Google Scholar 

  24. Jiang, Z. et al. eIF2alpha phosphorylation-dependent translation in CA1 pyramidal cells impairs hippocampal memory consolidation without affecting general translation. J. Neurosci. 30, 2582–2594 (2010).

    Article  CAS  Google Scholar 

  25. Sidrauski, C. et al. Pharmacological brake-release of mRNA translation enhances cognitive memory. eLife 2, e00498 (2013).

    Article  Google Scholar 

  26. Nadif Kasri, N., Nakano-Kobayashi, A. & Van Aelst, L. Rapid synthesis of the X-linked mental retardation protein OPHN1 mediates mGluR-dependent LTD through interaction with the endocytic machinery. Neuron 72, 300–315 (2011).

    Article  CAS  Google Scholar 

  27. Ron, D. & Harding, H.P. in Translational Control in Biology and Medicine Cold Spring Harbor vol. 39 (eds. Mathews, M.B., Sonenberg, N. & Hershey, J.W.B.) 345–368 (Cold Spring Harbor Laboratory Press, 2007).

  28. Harding, H.P. et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol. Cell 6, 1099–1108 (2000).

    Article  CAS  Google Scholar 

  29. Waung, M.W., Pfeiffer, B.E., Nosyreva, E.D., Ronesi, J.A. & Huber, K.M. Rapid translation of Arc/Arg3.1 selectively mediates mGluR-dependent LTD through persistent increases in AMPAR endocytosis rate. Neuron 59, 84–97 (2008).

    Article  CAS  Google Scholar 

  30. Park, S. et al. Elongation factor 2 and fragile X mental retardation protein control the dynamic translation of Arc/Arg3.1 essential for mGluR-LTD. Neuron 59, 70–83 (2008).

    Article  CAS  Google Scholar 

  31. Weiler, I.J. et al. Fragile X mental retardation protein is translated near synapses in response to neurotransmitter activation. Proc. Natl. Acad. Sci. USA 94, 5395–5400 (1997).

    Article  CAS  Google Scholar 

  32. Davidkova, G. & Carroll, R.C. Characterization of the role of microtubule-associated protein 1B in metabotropic glutamate receptor-mediated endocytosis of AMPA receptors in hippocampus. J. Neurosci. 27, 13273–13278 (2007).

    Article  CAS  Google Scholar 

  33. Graber, T.E. et al. Reactivation of stalled polyribosomes in synaptic plasticity. Proc. Natl. Acad. Sci. USA 110, 16205–16210 (2013).

    Article  CAS  Google Scholar 

  34. Sung, Y.J. et al. The fragile X mental retardation protein FMRP binds elongation factor 1A mRNA and negatively regulates its translation in vivo. J. Biol. Chem. 278, 15669–15678 (2003).

    Article  CAS  Google Scholar 

  35. Costa-Mattioli, M. et al. Translational control of hippocampal synaptic plasticity and memory by the eIF2alpha kinase GCN2. Nature 436, 1166–1173 (2005).

    Article  CAS  Google Scholar 

  36. Costa-Mattioli, M. et al. eIF2alpha phosphorylation bidirectionally regulates the switch from short- to long-term synaptic plasticity and memory. Cell 129, 195–206 (2007).

    Article  CAS  Google Scholar 

  37. Zhu, P.J. et al. Suppression of PKR promotes network excitability and enhanced cognition by interferon-gamma-mediated disinhibition. Cell 147, 1384–1396 (2011).

    Article  CAS  Google Scholar 

  38. Back, S.H. et al. Translation attenuation through eIF2alpha phosphorylation prevents oxidative stress and maintains the differentiated state in beta cells. Cell Metab. 10, 13–26 (2009).

    Article  CAS  Google Scholar 

  39. Stoica, L. et al. Selective pharmacogenetic inhibition of mammalian target of rapamycin complex I (mTORC1) blocks long-term synaptic plasticity and memory storage. Proc. Natl. Acad. Sci. USA 108, 3791–3796 (2011).

    Article  CAS  Google Scholar 

  40. Huang, W. et al. mTORC2 controls actin polymerization required for consolidation of long-term memory. Nat. Neurosci. 16, 441–448 (2013).

    Article  CAS  Google Scholar 

  41. Chan, J., Khan, S.N., Harvey, I., Merrick, W. & Pelletier, J. Eukaryotic protein synthesis inhibitors identified by comparison of cytotoxicity profiles. RNA 10, 528–543 (2004).

    Article  CAS  Google Scholar 

  42. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  Google Scholar 

  43. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    Article  CAS  Google Scholar 

  44. Schmidt, E.K., Clavarino, G., Ceppi, M. & Pierre, P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat. Methods 6, 275–277 (2009).

    Article  CAS  Google Scholar 

  45. Kaech, S. & Banker, G. Culturing hippocampal neurons. Nat. Protoc. 1, 2406–2415 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank K. Nakazawa and L. Van Aelst for the fPKR frozen embryos and Ophn1 shRNA, respectively. This work was supported by grants from the US National Institutes of Health to M.C.-M. (NIMH 096816, NINDS 076708) and R.J.K. (DK042394, DK088227, HL052173), the Intellectual Disability Research Center (P30HD024064) and Dan L. Duncan Cancer Center (P30CA125123) Genomic and RNA Profiling Cores, and the Cancer Prevention and Research Institute of Texas (CPRIT) RP100861.

Author information

Authors and Affiliations

Authors

Contributions

M.C.-M., G.V.D.P., W.H. and S.A.B. designed the experiments and wrote the manuscript; G.V.D.P. conducted electrophysiology and behavioral experiments and analyzed data; W.H. conducted behavioral, polysome profiling, qRT-PCR and immunoblotting experiments and analyzed data; S.A.B. conducted neuron culture, immunostaining, SUnSET and immunoblotting experiments and analyzed data; C.-C.H. performed firefly luciferase reporter experiments; P.B. analyzed RNA-seq data; A.P. contributed to the discussion of the electrophysiological experiments; C.S. and R.K. contributed to the characterization of ISRIB and the generation of Eif2s1A/A;ftg mice, respectively; K.K. and P.W. contributed to in-depth discussion of the project and editing of the manuscript.

Corresponding author

Correspondence to Mauro Costa-Mattioli.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Reduced eIF2α phosphorylation in the hippocampus of Eif2s1S/A mice, breeding strategy used to generate Eif2s1A/A;ftg mice and demonstration that DHPG elicited a normal mGluR-LTD in GFP-positive neurons expressed in WT mice.

a) Western blots show that compared to CA1 from WT Eif2s1S/S mice, in Eif2s1S/A mice, p-eIF2α is significantly reduced (n=3 mice, t=3.9, p=0.02). Results are displayed as mean ± SEM. Statistical significance was assessed by unpaired Student's t-test. b) Eif2s1S/S;ftg were first crossed to Eif2s1S/A mice. The resulting heterozygous transgenic (Eif2s1S/A;ftg) were then bred with Eif2s1S/A mice, which resulted in the generation of Eif2s1A/A;ftg. c) Application of DHPG (100mM, 5 min) elicited LTD in AAV-Cre-GFP+ neurons from WT mice (n=8 cells from 4 mice, t=11.38, p=9X10−6), indicating that the presence of GFP has no effect on LTD. Calibrations: 10 ms, 40 pA. Statistical significance was assessed by paired Student's t-test. Results are displayed as mean ± SEM.

Supplementary Figure 2 DHPG reduced surface GluR1 density in cultured hippocampal pyramidal neurons from WT mice but not in neurons from either Eif2s1S/A or Eif2s1A/A mice.

Surface GluR1 levels were detected as described in Methods. a-c) DHPG treatment (100mM, 5 min) reduced surface GluR1 density at 1 h post-treatment onset of cells from WT Eif2s1S/S (a) but not Eif2s1S/A (b) or Eif2s1A/A (c) mice. In contrast, synapsin-1 density was uniform among genotypes and treatment conditions. MAP2 immunostaining revealed that Eif2s1S/A and Eif2s1A/A cells displayed typical neuronal morphology in culture. Boxed areas indicate the dendritic segment shown in the expanded view immediately below the full-neuron, single-channel images. Merged images show combined data from the three individual channels. Scale bar indicates 20mm. d-e) Quantification of surface GluR1 (d) and synapsin-1 (e) density of DHPG-treated neurons normalized to vehicle-treated control neurons from the indicated genotype (n=51 for both Eif2s1S/S and Eif2s1S/A and n=34 for Eif2s1A/A; sGluR1, F(2,12)=19.3, p=0.00018; synapsin-1, F(2,12)=0.14, p=0.87). Statistical significance was assessed by a two-way ANOVA with Bonferroni correction for multiple comparisons. Results are displayed as mean ± SEM.

Supplementary Figure 3 Only when combined, sub-threshold concentrations of Sal003 and DHPG increased eIF2α phosphorylation in WT slices.

Separate applications of low concentrations of DHPG (10 μM, 5 min) or Sal003 (5 μM, 10 min) failed to induce eIF2α phosphorylation; but when at the same low concentrations Sal003 (5 μM, 10 min) immediately followed by DHPG (10 mM, 5 min) the phosphorylation of eIF2α was sharply boosted (n=4 independent experiments, F(3,20)=3.15, p=0.048). Statistical significance was assessed by a one-way ANOVA. Results are displayed as mean ± SEM.

Supplementary Figure 4 At a concentration that triggers phosphorylation of eIF2α, Sal003 elicited a sustained LTD in WT slices that was insensitive to mGluR1 and mGluR5 antagonists.

a) Brief application of Sal003 (20μM, 10 min) leads to a persistent long-lasting depression of evoked EPSCs (n=6 cells from 4 mice, t=7.07, p=0.0008, paired two sided t-test). b-c) The concurrent application (15min) of the mGluR1 blocker LY367385 (100mM) and the mGluR5 blocker MPEP (10 mM) prevents DHPG-induced LTD (b; n=6 cells from 2 mice, t=7.06,p=3.5X10-5, unpaired t-test) but had no effect on Sal003-induced LTD (c cells from 3 mice; n=8, t=0.53, p=0.61, unpaired two sided t-test). Calibrations: 10 ms, 40 pA. Results are displayed as mean ± SEM.

Supplementary Figure 5 Sal003 reduced surface GluR1 expression in control neurons, but not in eIF2α phosphorylation-deficient neurons.

a-c) One hour after starting application of Sal003 (20mM, 10 min), surface GluR1 density was reduced in neurons from WT Eif2s1S/S (a) but not Eif2s1S/A (b) or Eif2s1A/A (c) neurons. In contrast, synapsin-1 density did not differ significantly between genotypes or treatment conditions. MAP2 staining revealed that Eif2s1S/A and Eif2s1A/A cells displayed typical neuronal morphology. Boxed areas indicate the dendritic segment shown in the expanded view immediately below the full-neuron, single-channel images. Merged images show combined data from the three individual channels. Scale bar indicates 20mm. d-e) Quantification of Surface GluR1 (d) and synapsin-1 (e) density of Sal003-treated normalized to vehicle-treated control cultures from the indicated genotypes (n=55 for Eif2s1S/S and Eif2s1S/A and n=38 for Eif2s1A/A; d, p=0.0002; e, p=0.83). Statistical significance was assessed by two-way ANOVA with Bonferroni correction. Results are displayed as mean ± SEM.

Supplementary Figure 6 ISRIB, but not its inactive analog ISRIBinact, is a potent inhibitor of ATF4-driven translation.

a) Structures of ISRIB and its inactive analog, ISRIBinact. b) In HEK293T cells the ATF4 luciferase reporter (5'UTR of ATF4 mRNA fused to F-luciferase) was inhibited by ISRIB but not by the inactive analog ISRIBinact. Inhibition is plotted as function of the concentration of ISRIB or ISRIBinact. Inhibition assay was performed as described by Sidrauski et al.25.

Supplementary Figure 7 OPHN1 fluorescence intensity was positively correlated with p-eIF2α levels at individual synapses of cultured hippocampal neurons.

a-d) Representative two-channel immunostaining for p-eIF2α and OPHN1 from a dendritic segment in 14DIV control- (b), DHPG- (c), and Sal003-treated (d) hippocampal neuron cultures fixed 15 min after treatment onset. Framed areas (white squares) in (b) are expanded on the left (a). p-eIF2α and OPHN1 fluorescence intensity (FI) were positively correlated at individual synapses. b-d) As shown in the scatter plots depicting data from a total of 315 synapses from 9 neurons analyzed per condition. Lines in b-d represent the regression lines of the log-transformed population FI data (in AU). Slopes (m) and correlation coefficients (r) of the respective regression lines are indicated. Scale bars indicate 2mm. e-f) Bar graphs of the average FI of synaptic p-eIF2α (e) and OPHN1 (f) normalized to vehicle-treated controls [p-eIF2a: control = 1.0 ± 0.06, DHPG = 2.2 ± 0.1, Sal003 = 2.4 ± 0.07, F(2,24)=19.6, p=9X10-6; OPHN1: control = 1.0 ± 0.05, DHPG = 1.9 ± 0.08, Sal003 = 1.8 ±0.1, F(2,24)=21.0, p=5.4X10-6]. Statistical significance was assessed by one-way ANOVA. Results are displayed as mean ± SEM.

Supplementary Figure 8 OPHN1 and Arc mRNA translation are required for mGluR-LTD in CA1.

a) A specific shRNA against OPHN1 mRNA blocked DHPG-induced LTD (n=6 cells from 5 mice, t=0.88, p=0.42) whereas scrambled control shRNA did not (n=5 cells from 3 mice, t=14.4, p=0.00014; group difference t=6.28, p=0.0002). Viruses expressing GFP together with OPHN1-shRNA or scrambled-control-shRNA were injected into the hippocampus of WT mice. Examples of inset traces from which plots were drawn are illustrated. b) DHPG-induced LTD was suppressed by an antisense oligo (as) that blocks Arc synthesis (n=12 cells from 5 mice, t=0.77, p=0.46) but not by the control mismatch oligo (ms) (n=8 cells from 5 mice, t=6.61, p=0.0003; group difference t=6.28, p=2X10−5). Calibrations: 10 ms, 40 pA. c) Sal003 increased eIF2α phosphorylation, but not Arc levels in hippocampal slices (n=5 independent experiments, t=0.5, p=0.62). Statistical significance was assessed by paired Student's t-test. Results are displayed as mean ± SEM.

Supplementary Figure 9 Spatial recognition increased eIF2α phosphorylation and OPHN1 levels in the hippocampus and the exploratory behavior was similar in all experimental groups.

a,c) Western blots from hippocampal tissue show increased eIF2α phosphorylation (a) and OPHN1 levels (c) after exposure to two objects on day 2 (n=3 mice). b,d) Quantification of normalized p-eIF2α (b, at 10 min t=2.91, p=0.033; at 90 min t=3.37, p=0.020) and OPHN1 (d, at 10 min t=0.287, p=0.79; at 90 min t=2.98, p=0.031). e-g) Raw data for individual animals during training (day 2) or testing (day 3). Plot shows exploration times for individual Eif2s1S/S and Eif2s1S/A mice (e), vehicle-injected and ISRIB-injected mice (f) and control-shRNA-injected and OPHN1-shRNA-injected mice (g) on Day 2 and 3. Statistical significance was assessed by unpaired Student's t-test. Results are displayed as mean ± SEM.

Supplementary Figure 10 Images of full-length blots presented in all figures.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 (PDF 23068 kb)

Supplementary Methods Checklist (PDF 622 kb)

Supplementary Table 1

mRNAs translationally upregulated by mGluR activation. (XLSX 28 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Di Prisco, G., Huang, W., Buffington, S. et al. Translational control of mGluR-dependent long-term depression and object-place learning by eIF2α. Nat Neurosci 17, 1073–1082 (2014). https://doi.org/10.1038/nn.3754

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3754

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing