Volume 25, Issue 4, Pages e4 (October 2018)

Slides:



Advertisements
Similar presentations
The chemokine CCL2 activates p38 mitogen-activated protein kinase pathway in cultured rat hippocampal cells  Jungsook Cho, Donna L. Gruol  Journal of.
Advertisements

Volume 70, Issue 4, Pages (May 2011)
Volume 77, Issue 2, Pages (January 2013)
Volume 49, Issue 4, Pages (February 2006)
Efficient, Complete Deletion of Synaptic Proteins using CRISPR
Volume 22, Issue 4, Pages (April 1999)
Memory Enhancement by Targeting Cdk5 Regulation of NR2B
Volume 54, Issue 6, Pages (June 2007)
Volume 40, Issue 4, Pages (November 2003)
Zinc Dynamics and Action at Excitatory Synapses
Volume 44, Issue 3, Pages (October 2004)
Distinct NMDA Receptors Provide Differential Modes of Transmission at Mossy Fiber- Interneuron Synapses  Saobo Lei, Chris J McBain  Neuron  Volume 33,
NMDA Receptor-Dependent LTD Requires Transient Synaptic Incorporation of Ca2+- Permeable AMPARs Mediated by AKAP150-Anchored PKA and Calcineurin  Jennifer L.
NMDA Induces Long-Term Synaptic Depression and Dephosphorylation of the GluR1 Subunit of AMPA Receptors in Hippocampus  Hey-Kyoung Lee, Kimihiko Kameyama,
DAPK1 Mediates LTD by Making CaMKII/GluN2B Binding LTP Specific
Volume 80, Issue 4, Pages (November 2013)
Alternative N-Terminal Domains of PSD-95 and SAP97 Govern Activity-Dependent Regulation of Synaptic AMPA Receptor Function  Oliver M. Schlüter, Weifeng.
LTP Requires a Unique Postsynaptic SNARE Fusion Machinery
Volume 86, Issue 2, Pages (April 2015)
Volume 56, Issue 4, Pages (November 2007)
Volume 70, Issue 2, Pages (April 2011)
Andres Barria, Roberto Malinow  Neuron 
Hippocampus and Entorhinal Cortex Recruit Cholinergic and NMDA Receptors Separately to Generate Hippocampal Theta Oscillations  Zhenglin Gu, Georgia M.
Volume 11, Issue 12, Pages (June 2015)
Volume 74, Issue 3, Pages (May 2012)
Tumor Necrosis Factor-α Mediates One Component of Competitive, Experience- Dependent Plasticity in Developing Visual Cortex  Megumi Kaneko, David Stellwagen,
Volume 77, Issue 2, Pages (January 2013)
The Environment versus Genetics in Controlling the Contribution of MAP Kinases to Synaptic Plasticity  Shaomin Li, Xuejun Tian, Dean M. Hartley, Larry.
The Retromer Supports AMPA Receptor Trafficking During LTP
Volume 6, Issue 3, Pages (February 2014)
A Cooperative Mechanism Involving Ca2+-Permeable AMPA Receptors and Retrograde Activation of GABAB Receptors in Interpeduncular Nucleus Plasticity  Peter.
A Role for Stargazin in Experience-Dependent Plasticity
Volume 68, Issue 5, Pages (December 2010)
Subunit-Specific NMDA Receptor Trafficking to Synapses
Volume 52, Issue 2, Pages (October 2006)
Volume 92, Issue 1, Pages (October 2016)
Volume 22, Issue 10, Pages (March 2018)
Experience-Dependent Equilibration of AMPAR-Mediated Synaptic Transmission during the Critical Period  Kyung-Seok Han, Samuel F. Cooke, Weifeng Xu  Cell.
Volume 57, Issue 2, Pages (January 2008)
CAPS-1 and CAPS-2 Are Essential Synaptic Vesicle Priming Proteins
Volume 20, Issue 6, Pages (August 2017)
Barrel Cortex Critical Period Plasticity Is Independent of Changes in NMDA Receptor Subunit Composition  Hui-Chen Lu, Ernesto Gonzalez, Michael C Crair 
Volume 16, Issue 5, Pages (August 2016)
Volume 82, Issue 1, Pages (April 2014)
Rui Qin, Shuai Cao, Tianjie Lyu, Cai Qi, Weiguang Zhang, Yun Wang 
Volume 71, Issue 6, Pages (September 2011)
Volume 62, Issue 2, Pages (April 2009)
Bo Li, Ran-Sook Woo, Lin Mei, Roberto Malinow  Neuron 
Volume 25, Issue 1, Pages e6 (October 2018)
Volume 22, Issue 9, Pages (February 2018)
PKA-GluA1 Coupling via AKAP5 Controls AMPA Receptor Phosphorylation and Cell- Surface Targeting during Bidirectional Homeostatic Plasticity  Graham H.
Volume 18, Issue 12, Pages (March 2017)
Volume 3, Issue 3, Pages (March 2013)
Volume 23, Issue 4, Pages (April 2018)
Hippocalcin Functions as a Calcium Sensor in Hippocampal LTD
Volume 20, Issue 8, Pages (August 2017)
Volume 21, Issue 1, Pages (October 2017)
Volume 135, Issue 3, Pages (October 2008)
Volume 51, Issue 4, Pages (August 2006)
Yanghong Meng, Yu Zhang, Zhengping Jia  Neuron 
Volume 24, Issue 6, Pages (August 2018)
Genetic Dissection of Presynaptic and Postsynaptic BDNF-TrkB Signaling in Synaptic Efficacy of CA3-CA1 Synapses  Pei-Yi Lin, Ege T. Kavalali, Lisa M.
Volume 71, Issue 6, Pages (September 2011)
Social Isolation Induces Rac1-Dependent Forgetting of Social Memory
Pernille Bülow, T.J. Murphy, Gary J. Bassell, Peter Wenner 
Volume 9, Issue 5, Pages (December 2014)
Cary Soares, Kevin F.H. Lee, Jean-Claude Béïque  Cell Reports 
Volume 26, Issue 8, Pages e6 (February 2019)
SAP102 Mediates Synaptic Clearance of NMDA Receptors
Postsynaptic Complexin Controls AMPA Receptor Exocytosis during LTP
Presentation transcript:

Volume 25, Issue 4, Pages 841-851.e4 (October 2018) The Developmental Shift of NMDA Receptor Composition Proceeds Independently of GluN2 Subunit-Specific GluN2 C-Terminal Sequences  Sean McKay, Tomás J. Ryan, Jamie McQueen, Tim Indersmitten, Katie F.M. Marwick, Philip Hasel, Maksym V. Kopanitsa, Paul S. Baxter, Marc-André Martel, Peter C. Kind, David J.A. Wyllie, Thomas J. O’Dell, Seth G.N. Grant, Giles E. Hardingham, Noboru H. Komiyama  Cell Reports  Volume 25, Issue 4, Pages 841-851.e4 (October 2018) DOI: 10.1016/j.celrep.2018.09.089 Copyright © 2018 The Author(s) Terms and Conditions

Cell Reports 2018 25, 841-851.e4DOI: (10.1016/j.celrep.2018.09.089) Copyright © 2018 The Author(s) Terms and Conditions

Figure 1 The GluN2B CaMKII Site Is Dispensable for the Developmental Switch In Vitro (A) Schematic of the GluN2BΔCaMKII amino acid changes. (B and C) Altered GluN2B phosphorylation in GluN2BΔCaMKII/ΔCaMKII neurons. Cortical neuronal extracts were prepared and analyzed by western blot with the indicated antibodies, normalized to total GluN2B. (B) shows quantitation (mean ± SEM here and throughout), and (C) shows an example. ∗p < 0.05 versus GluN2B+/+ (n = 4). (D) NMDAR current density at the indicated ages for GluN2BWT/WT and GluN2BΔCaMKII/ΔCaMKII neurons. ∗p < 0.05 versus DIV7 or DIV8 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (GluN2BWT/WT: n = 14 [DIV7 or DIV8], n = 18 [DIV14–18]; GluN2BΔCaMKII/ΔCaMKII: n = 15 [DIV7 or DIV8], n = 17 [DIV14–18]). (E) Extrasynaptic NMDAR currents were measured in GluN2BWT/WT and GluN2BΔCaMKII/ΔCaMKII neurons (n = 8 per genotype). (F and G) GluN2A and GluN2B expression analyzed by western blot at DIV8 and DIV16 for GluN2BWT/WT and GluN2BΔCaMKII/ΔCaMKII neurons. (F) shows quantitation and (G) shows an example. ∗p < 0.05 versus DIV8 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (n = 8). (H and I) Neocortices of GluN2BWT/WT and GluN2BΔCaMKII/ΔCaMKII mice were subjected to immunoprecipitation with an antibody to the N terminus of GluN2B, followed by analysis of GluN2B and CaMKIIα content. (H) shows an example and (I) shows quantitation. ∗p < 0.05; n = 8 per genotype. (J) Percentage blockade of NMDAR currents by ifenprodil (3 μM) at the indicated stages for GluN2BWT/WT and GluN2BΔCaMKII/ΔCaMKII neurons. ∗p < 0.05 versus DIV7 or DIV8 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (GluN2BWT/WT: n = 14 [DIV7 or DIV8], n = 18 [DIV14–18]; GluN2BΔCaMKII/ΔCaMKII: n = 15 [DIV7 or DIV8], n = 17 [DIV14–18]). (K and L) Percentage potentiation of NMDAR currents by 100 μM spermine was measured at the indicated stages for GluN2BWT/WT and GluN2BΔCaMKII/ΔCaMKII neurons. ∗p < 0.05 versus DIV7 or DIV8 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (GluN2BWT/WT: n = 13 [DIV7 or DIV8], n = 18 [DIV14–18]; GluN2BΔCaMKII/ΔCaMKII: n = 18 [DIV7 or DIV8], n = 14 [DIV14–18]). (K) shows quantitation and (L) shows example traces; scale bar: 200 pA/2 s. (M and N) Neocortical extracts from P14 and P28 mice were analyzed for GluN2A expression, normalized to β-actin. (M) shows quantitation and (N) shows an example. ∗p < 0.05 versus P14 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (n = 8). (O and P) Post-synaptic density (PSD) extracts from P14 and P28 mice of the indicated genotype were analyzed for GluN2A and GluN2B expression, normalized to β-actin, and the ratio calculated. (O) shows quantitation and (P) shows an example. ∗p < 0.05 versus P14 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (n = 8). Cell Reports 2018 25, 841-851.e4DOI: (10.1016/j.celrep.2018.09.089) Copyright © 2018 The Author(s) Terms and Conditions

Figure 2 The GluN2B CaMKII Binding Site Is Dispensable for Theta Burst LTP (A–C) Activity-dependent signaling to ERK1/2 does not require GluN2B CTD-specific sequences. DIV9 cortical neurons of the indicated genotypes were treated with TTX (500 nM), KN-62 (10 μM), or MK-801 (10 μM) for 1 hr, after which protein extracts were made and subjected to western blot analysis for phospho-ERK1/2 levels, normalized to total ERK1/2. (A) shows quantitation and (B) and (C) show example blots.∗p < 0.05 two-way ANOVA plus Dunnett’s post hoc test. No genotype-dependent effects were observed (p = 0.39) or genotype-drug interactions (p = 0.16; n = 8 GluN2BWT/WT; n = 4 GluN2BΔCaMKII/ΔCaMKII; n = 4 GluN2B2A(CTR)/2A(CTR)). (D–F) Cumulative probability plots show amplitude (D) and frequency (E) of miniature EPSCs in GluN2BWT/WT and GluN2BΔCaMKII/ΔCaMKII slices. Results are from 11 cells from 3 GluN2BWT/WT mice and 13 cells from 3 GluN2BΔCaMKII/ΔCaMKII mice; p = 0.75 (amplitude); p = 0.94 (frequency); unpaired t test. (F) shows example traces. Scale bar: 10 pA/100 ms. (G) NMDAR:AMPAR EPSC ratios at −80 and +40 mV was measured in GluN2BWT/WT (n = 25; 4 mice) and GluN2BΔCaMKII/ΔCaMKII (n = 30; 4 mice) neurons (unpaired t test; p = 0.95). (H) Weighted time constant (τw) for the decay of EPSCs recorded in (A) at +40 mV was determined using double exponential fits (unpaired t test; p = 0.63). (I) Example traces. Scale bar: 100 pA/20 ms. (J and K) Theta-burst stimulation elicited pathway-specific LTP of synaptic transmission in hippocampal CA1 area. Normalized magnitude of this potentiation 60–65 min after LTP induction did not differ significantly in GluN2BWT/WT mice (29 slices; n = 9) compared to GluN2BΔCaMKII/ΔCaMKII (35 slices; n = 11); p = 0.162 (two-way nested ANOVA). (J) shows quantitation of data and (K) shows example traces before and after LTP induction. (K) Traces show example fEPSP traces immediately before and 1 hr after theta-burst stimulation. Scale bar: 0.5 mV/2 ms. Cell Reports 2018 25, 841-851.e4DOI: (10.1016/j.celrep.2018.09.089) Copyright © 2018 The Author(s) Terms and Conditions

Figure 3 Distinct GluN2 CTDs Are Not Required for the 2B-to-2A Switch (A) Schematic illustrating the C-terminal domain exchange in the GluN2A2B(CTR)/2B(CTR) mouse. (B) NMDAR current density in GluN2AWT/WT (n = 30) and GluN2A2B(CTR)/2B(CTR) (n = 24) neurons, recorded DIV14–18. (C and D) Percentage potentiation of NMDAR currents by 100 μM spermine was measured in GluN2AWT/WT and GluN2A2B(CTR)/2B(CTR) neurons. ∗p < 0.05 versus DIV7 or DIV8 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (GluN2AWT/WT: n = 19 [DIV7 or DIV8], n = 18 [DIV14–18]; GluN2A2B(CTR)/2B(CTR): n = 16 [DIV7 or DIV8], n = 17 [DIV14–18]). (C) shows quantitation of data and (D) shows example traces; scale bar: 200 pA/2 s. (E and F) Neocortical extracts from P14 and P28 mice of the indicated genotype were analyzed for GluN2A expression, normalized to β-actin. ∗p < 0.05 versus P14 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (n = 8). (E) shows quantitation and (F) shows example blots. (G and H) PSD extracts from P14 and P28 mice of the indicated genotype were analyzed for GluN2A and GluN2B expression, normalized to β-actin, and the ratio calculated and scaled such that the ratio at P14 for WT = 1. ∗p < 0.05 versus P14 of the same genotype; two-way ANOVA plus Sidak’s post hoc test (n = 8). (G) shows quantitation and (H) shows an example blot. Cell Reports 2018 25, 841-851.e4DOI: (10.1016/j.celrep.2018.09.089) Copyright © 2018 The Author(s) Terms and Conditions

Figure 4 GluN2A Expression Is Sufficient to Displace GluN2B from NMDARs (A–D) Total GluN2B expression was measured in extracts from the neocortex at the indicated stages for the indicated genotypes (n = 8 per genotype). (A) and (B) show quantitation and example blots, respectively, for GluN2AWT/WT versus GluN2A2B(CTR)/2B(CTR). (C) and (D) show quantitation and example blots, respectively, for GluN2AWT/WT vs. GluN2BΔCaMKII/ΔCaMKII. (E–G) Ectopic GluN2A expression is sufficient to displace GluN2B from NMDARs. Young mouse (E and F) and rat (G) neurons at DIV7 were transfected with a control (β-globin) or GluN2A-encoding vector and spermine (100 μM) potentiation of NMDAR currents measured 72 hr later. ∗p < 0.05 (F: n = 10 of both condition; G: n = 8 control; n = 10 GluN2A). (H) Percentage potentiation of NMDAR currents by spermine (200 μM) in DIV15 or DIV16 rat cortical neurons was measured at the indicated stages for GluN2A+/+, GluN2A+/−, and GluN2A−/− genotypes. Person r correlation coefficient: −0.9954; p = 0.031 (one-tailed test); n = 12 cells per genotype. (I) Western blot illustrating GluN2A expression in GluN2A+/+, GluN2A+/–, and GluN2A–/– neurons. (I) A western blot confirming the absence of GluN2A expression in GluN2A−/− and an intermediate expression level in GluN2A+/− neurons. Cell Reports 2018 25, 841-851.e4DOI: (10.1016/j.celrep.2018.09.089) Copyright © 2018 The Author(s) Terms and Conditions