Supplementary Figure 1. Screening for monoclonal antibodies against GluA1 by immunoblotting.

Similar documents
Stargazin regulates AMPA receptor trafficking through adaptor protein. complexes during long term depression

Supplemental figures (1-9) Gu et al. ADF/Cofilin-Mediated Actin Dynamics Regulate AMPA Receptor Trafficking during Synaptic Plasticity

Nature Neuroscience: doi: /nn Supplementary Figure 1

Supplementary Figure 1. APP cleavage assay. HEK293 cells were transfected with various

LiveReceptor AMPAR <Endogenous AMPAR Labeling Reagent>

DCLK-immunopositive. Bars, 100 µm for B, 50 µm for C.

Supplementary Figures and Legends.

Acoustically targeted chemogenetics for the non-invasive control of neural circuits

Supplementary Figure 1. α-synuclein is truncated in PD and LBD brains. Nature Structural & Molecular Biology: doi: /nsmb.

To examine the silencing effects of Celsr3 shrna, we co transfected 293T cells with expression

Supplementary Figure 1. Serial deletion mutants of BLITz.

Supplementary Figures and supplementary figure legends

Nature Neuroscience: doi: /nn Supplementary Figure 1

Figure S1 is related to Figure 1B, showing more details of outer segment of

Supplementary Figure 1. Confirmation of sirna in PC3 and H1299 cells PC3 (a) and H1299 (b) cells were transfected with sirna oligonucleotides

Supplementary Figure 1. Drawing of spinal cord open-book preparations and DiI tracing. Nature Neuroscience: doi: /nn.3893

Supplementary Figure 1. Antigens generated for mab development (a) K9M1P1-mIgG and hgh-k9m1p1 antigen (~37 kda) expression verified by western blot

Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in

SUPPLEMENTARY FIGURES

Parthanatos mediates AIMP2-activated age-dependent dopaminergic neuronal loss

Supplementary Figure 1 Collision-induced dissociation (CID) mass spectra of peptides from PPK1, PPK2, PPK3 and PPK4 respectively.

Supplementary Figure 1. Pathologically phosphorylated Tau is localized to the presynaptic terminals of Alzheimer s disease (AD) patient brains.

Class 7: Methods in Research By: Ray

T H E J O U R N A L O F C E L L B I O L O G Y

T H E J O U R N A L O F C E L L B I O L O G Y

DRG Pituitary Cerebral Cortex

Supplementary Information

Supplementary Information Cyclin Y inhibits plasticity-induced AMPA receptor exocytosis and LTP

Supplementary Information

SUPPLEMENTARY FIGURE 1

Supplementary Figure 1

Genetically targeted all-optical electrophysiology with a transgenic Credependent

Supplementary information

Regulation of Acetylcholine Receptor Clustering by ADF/Cofilin- Directed Vesicular Trafficking

Supplementary Information

Figure S1. USP-46 is expressed in several tissues including the nervous system

Supplementary Figure 1: Expression of RNF8, HERC2 and NEURL4 in the cerebellum and knockdown of RNF8 by RNAi (a) Lysates of the cerebellum from rat

Serine phosphorylation of ephrinb2 regulates trafficking of synaptic

Regulation of axonal and dendritic growth by the extracellular calcium-sensing

Disrupted-in-Schizophrenia-1 (DISC1) regulates spines of the glutamate synapse via Rac1

SUPPLEMENTARY INFORMATION

Supplementary Figure1: ClustalW comparison between Tll, Dsf and NR2E1.

Supplementary Figure 1. Generation of B2M -/- ESCs. Nature Biotechnology: doi: /nbt.3860

Supplementary Figure 1. ERK signaling is not activated at early hypertension. a, Western blot analysis for the level of phospho-erk (perk) and total

Supporting Information

Supplementary Figure 1. The integrity of myelin sheaths is not affected in MAP6 KO mice

Supplementary Figure 1.

Methanol fixation allows better visualization of Kal7. To compare methods for

CD93 and dystroglycan cooperation in human endothelial cell adhesion and migration

Supplementary Figure 1. Characterization of EVs (a) Phase-contrast electron microscopy was used to visualize resuspended EV pellets.

Control of cortex development by ULK4, a rare risk gene for mental disorders including schizophrenia

Figure legends for supplement

SUPPLEMENTARY INFORMATION SUPPLEMENTARY FIGURES

In vitro EGFP-CALI comprehensive analysis. Liang CAI. David Humphrey. Jessica Wilson. Ken Jacobson. Dept. of Cell and Developmental Biology

Supplementary Figure 1. Effects of STOML3-modulating molecules on other stomatin-domain proteins

0.9 5 H M L E R -C tr l in T w is t1 C M

Supplementary Information attached to:

Single cell resolution in vivo imaging of DNA damage following PARP inhibition. Supplementary Data

(B) Comparable expression of major integrin subunits and glycoproteins on the surface of resting WT and Lnk -/- platelets.

Supplementary Figure 1 Phosphorylated tau accumulates in Nrf2 (-/-) mice. Hippocampal tissues obtained from Nrf2 (-/-) (10 months old, 4 male; 2

Supplementary Information. Real-time imaging of oxidative and nitrosative stress in the liver of live animals for drug-toxicity testing

Expanded View Figures

Farnesoid X Receptor and its ligands inhibit the function of platelets

Supplementary Figure 1. The Hsp70 acetylation level is related to the co-chaperone binding of Hsp70 under various stress conditions.

Regulation of hepcidin expression by inflammation-induced activin B

Nature Biotechnology: doi: /nbt Supplementary Figure 1. Design and sequence of system 1 for targeted demethylation.

Reporting Checklist for Nature Neuroscience

Supplementary Fig. 1 related to Fig. 1 Clinical relevance of lncrna candidate

Supplementary Fig. 1

Supplementary Fig.1 Luton

Post-expansion antibody delivery, after epitope-preserving homogenization.

(A) Schematic illustration of sciatic nerve ligation. P, proximal; D, distal to the ligation site.

Expanded View Figures

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION

Expanded View Figures

Supplemental Figure 1 Human REEP family of proteins can be divided into two distinct subfamilies. Residues (single letter amino acid code) identical

Electronic Supplementary Information

supplementary information

Reporting Checklist for Nature Neuroscience

Cancer cells that survive radiation therapy acquire HIF-1 activity and translocate toward tumor blood vessels Supplementary Information

Supplemental Methods. Animals

Assembly of synapses by neuronal adhesion molecules: single molecule studies

Nature Medicine: doi: /nm.4356

Supporting Figure 1: Meis2 and Pax6 transcripts in the adult murine brain detected by in situ

Gene Sequence Fragment size ΔEGFR F 5' GGGCTCTGGAGGAAAAGAAAG GT 3' 116 bp R 5' CTTCTTACACTTGCGGACGC 3'

Supplementary Figure S1. N-terminal fragments of LRRK1 bind to Grb2.

In vivo recording, forepaw denervation, and isolation of slices: Methods for mapping the forepaw/lower jaw border in anesthetized adult rat primary

Improved Immunoblot by Brief Application of Low Dose Paraformaldehyde

Nature Biotechnology: doi: /nbt Supplementary Figure 1

Direct Imaging of APP Proteolysis in Living Cells

SUPPLEMENTARY INFORMATION

Nature Medicine: doi: /nm.4169

Regulation of Synaptic Structure and Function by FMRP- Associated MicroRNAs mir-125b and mir-132

Sélection Internationale Ens Ulm 2012, Cell Biology

A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer s disease

Intestinal Epithelial Cell-Specific Deletion of PLD2 Alleviates DSS-Induced Colitis by. Regulating Occludin

VERIFY Tagged Antigen. Validation Data

by Neurobasal medium (supplemented with B27, 0.5mM glutamine, and 100 U/mL

Grb2-Mediated Alteration in the Trafficking of AβPP: Insights from Grb2-AICD Interaction

Transcription:

Supplementary Figure 1 Screening for monoclonal antibodies against GluA1 by immunoblotting. Hippocampal extract was subjected to western blotting with the hybridoma supernatants of candidate monoclonal antibodies against GluA1. Positive antibodies are labeled with an asterisk. First upper left lane was blotted with an anti- GluA1 monoclonal antibody (Millipore, C3T, raised against the GluA1 cytoplasmic tail: anti-glua1-ct). Second upper left lane was blotted with an anti-gfp monoclonal antibody (anti-gfp). To check the weak positive samples, we also examined Z9131 and Z9141 in staining shown in figure 1c. To test the negative samples, we also examined Z9143-45 in figure 1c.

Supplementary Figure 2 Basic conditions of the CALI experiment with Z9139-eosin in vitro and effect of NASPM. a. Power and time dependence of light irradiation on the CALI effect with Z9139-eosin in vitro. Glutamate puff-induced responses were obtained in GluA1 expressing CHO cells with the indicated combinations of time and power for CALI. The data for 0 mw and 7.5 mw/cm 2, 1 min were the same as in Figure 2a. F(4,31)=5.618, *p<0.001, **p<0.05 and p=0.296 in 2.5mW/cm 2-1min vs 0mW (one-way ANOVA followed by post hoc analysis with the Fisher s PLSD test). n=5, 7, 6, 8 and 8 cells for 0mW, 2.5mW-1min, 5mW-1min, 7.5mW-1min and 7.5mW-0.5min, respectively. b. A saturating concentration of NASPM reduced the synaptic AMPA receptor-mediated currents in hippocampal primary cultures ~50%. AMPA receptor-mediated synaptic currents after NASPM application at different concentrations; values are the ratio of the current after to before NASPM application. Note that the effect of a saturating concentration (20 M) of NASPM on synaptic AMPA receptor currents was examined in the experiment in Figure 3d. F(5,49)=9.805, *p<0.001, **p<0.01 and p=0.273 in 0.01 vs 0 M (one-way ANOVA followed by post hoc analysis with the Fisher s PLSD test). n=11, 7, 8, 11, 9 and 9 cells for 0 M, 0.01 M, 0.1 M, 0.5 M, 5 M and 20 M, respectively. n.s. indicates no significance.

Supplementary Figure 3 Endosomal trafficking is not affected by CALI with Z9139-eosin. Fluorescence images of Alexa488-transferrin before and after recycling for 30 min with or without CALI using Z9139- eosin. The relative fluorescence intensity was examined before and after recycling in the dendrites and cell body (n=30 regions of interest [ROIs]). ROIs were obtained from 3 independent experiments. Scale bar indicates 5 m. t(52.769)=1.002, p=0.321 (unpaired two-tailed t-test). n.s. indicates no significance.

Supplementary Figure 4 Basic properties of the IA task-related experiment. a. Expression of GluA1 cytoplasmic tail in the hippocampus prevents the acquisition of IA memory. Latency period for re-entering the dark box of IA-trained mice expressing GFP or GFP-ctail (cytoplasmic terminus of GluA1: a peptide that blocks synaptic GluA1 delivery) in the CA1 region of the dorsal hippocampus. The latency period for re-entering the dark box was significantly shorter in the GFP-ctail-expressing animals than in the GFP-expressing ones. *p<0.001 (Mann-Whitney U test). n=9 and 7 animals for GFP and c-tail, respectively. b. Membrane properties of the examined neurons (n=14 cells shown in Figure 4b) in slices with or without IA conditioning. t(26)=1.127, p =0.270 in Cm, t(26)=0.028, p =0.781 in Rm and t(26)=0.139, p =0.891 in Ra (unpaired two-tailed t-test). n.s. indicates no significance.

Supplementary Figure 5 Infusion of Z9139 into the hippocampus in vivo. a. (Left) Bright field image of a brain slice including the hippocampus obtained from an animal in which a cannula was implanted. (Right) Location of the cannula labeled with DiI. b. Distribution of injected Alexa546-labeled Z9139 in the hippocampus of mice in which a cannula was implanted. To visualize the Z9139 antibody, it was labeled with Alexa546. Z9139-Alexa546 was bilaterally injected at the same stereotaxic coordinates. After 7.5 hours (the same duration as in the in vivo CALI experiment), the mice were fixed by perfusion, and the brain was removed and sliced (100 m) on a microtome. The coordinates 900-1200 m are the distance from the anterior end of the hippocampus. The intensity of the Alexa-546 signal was plotted along the CA1 region (yellow dashed line) in the right panels. Scale bar indicates 200 m. The Z9139 antibody diffused approximately 400 m laterally and 200 m sagittally in the CA1 region. c. Quantitative analysis of the alexa546-labeled Z9139 diffusion using the slices at 1000 and 1100 m obtained from 4 animals. d. Location of whole-cell recordings after in vivo CALI. Right panel is an enlarged view of the red box in the left panel. Yellow box indicates the position of the light cannula, determined by the injury in the cortex. We performed whole-cell patch clamp experiments (Figures 4e, 5b) within the white box, which was 150 m bilaterally, from just under the cannula (asterisk) along the CA1 region. Scale bars are 1 mm (left panel) and 150 m (right panel).

Supplementary Figure 6 in vivo CALI with Z9139-eosin under different conditions. a. Power and time dependence of the laser irradiation on the in vivo CALI effect. Latency for re-entering the dark box after the IA task in Z9139-eosin-treated animals with CALI was measured under the indicated conditions. The data for 0 mw and 60 mw, 2 min were same as in Figure 4c. *p<0.005, p=0.188 in 60mW-1min and p=0.854 in 30mW-2min vs 0mW (Kraskal-Wails test by post hoc analysis with Dunne s test). n=6, 8, 6 and 10 animals for 0mW, 60mW-1min, 30mW-2min and 60mW-2min, respectively. n.s. indicates no significance. b. The in vivo CALI effect with Z9139-eosin was retained for 1 week. The latency period for re-entering the dark box of IA-trained mice 1 week after treatment with or without CALI by Z9139-eosin. *p<0.05 (Mann-Whitney U test). n=11 and 15 animals for CALI- and CALI+, respectively. c. In vivo CALI with Z9139-eosin induced fear memory erasure in 8-week-old ICR mice. Eight-week-old ICR mice were subjected to in vivo CALI with Z9139-eosin. The experimental conditions were same as those used for 4-week-old ICR mice, shown in Figure 4c. *p<0.005 in Z9139 and p=0.852 in anti-myc (Mann-Whitney U test). n=6, 8, 7 nad 8 animals for Myc CALI-, Myc CALI+, Z9139 CALI- and Z9139 CALI+, respectively. n.s. indicates no significance.

Supplementary Figure 7 In vivo CALI with Z9139-eosin did not induce acute damage in memory-erased animals. Acute effect of in vivo CALI with Z9139-eosin was examined by re-conditioning 30 min after in vivo CALI. We reconditioned the mice whose IA memory we had erased by in vivo CALI with Z9139-eosin (IA test 1 & re-cond.) and tested them by IA test 2. Latencies for re-entering the dark box after the IA task in Z9139-eosin-treated animals without or with (IA test 1) CALI and in mice subjected to re-conditioning after Z9139-CALI (IA test 2). n=5 (CALI-) and n=8 (IA test1 and test2) animals. Data were analyzed by Kraskal-Wails test by post hoc analysis with Dunne s test. *p<0.01 vs CALI- and p=0.352 in IA test2 vs CALI-. n.s. indicates no significance.

Supplementary Figure 8 Specificity of in vivo CALI with Z9139-eosin for GluA1 homomeric receptors. a. GluA1 homomers were not detectable in synapses before learning. Responses at the hippocampal CA3-CA1 synapses in the acute brain slices obtained from IA-trained or untrained animals with or without NASPM. Average AMPA/NMDA ratio with or without NASPM before and after learning. t(18)=2.662, *p<0.05 in 1hr after learning and t(14)=0.667, p=0.515 in before learning (unpaired two-tailed t-test). n=8, 8, 9 and 11 cells for before learning DW, before learning NASPM, after learning DW and after learning NASPM, respectively. b. In vivo CALI with Z9139-eosin did not inactivate the GluA1 homomer-unrelated AMPA response. Responses at hippocampal CA3-CA1 synapses in the acute brain slices obtained from IA-untrained animals with or without in vivo CALI. Average AMPA/NMDA ratio with or without in vivo CALI. t(12)=0.388, p=0.705 (unpaired two-tailed t-test). n=8 cells, respectively. In Supplementary Figure 8a, the results of DW(before learning), NASPM(before learning), DW(after learning) and NASPM(after learning) were obtained from 6, 3, 5 and 3 animals, respectively. In Supplementary Figure 8b, the results of CALI- and CALI+ were obtained from 4 animals, respectively. n.s. indicates no significance.

Supplementary Figure 9 Re-learning of the IA task increased the synaptic delivery of homomeric GluA1. Synaptic responses at the hippocampal CA3-CA1 synapses in acute brain slices obtained from animals with or without reconditioning 0.5 h or 24 h after in vivo CALI. Average rectification index at hippocampal CA3-CA1 synapses with or without re-conditioning after in vivo CALI. t(14)=2.650, *p<0.05 in 24hr and t(15)=1.427, p=0.174 in 0.5hr (unpaired twotailed t-test). n=8, 9, 7 and 9 cells for 0.5hr re-cond-, 0.5hr re-cond+, 24hr re-cond- and 24hr re-cond+, respectively. The results of 0.5hr(re-cond-), 0.5hr(re-cond+), 24hr(re-cond-) and 24hr(re-cond+) were obtained from 3, 4, 4 and 3 animals, respectively. n.s. indicates no significance.

Supplementary Figure 10 Spatial specificity of in vivo CALI with Z9139-eosin. a. Expression of GluA1 cytoplasmic tail in the auditory cortex did not prevent the acquisition of IA memory. Latency period for re-entering the dark box of IA-trained mice expressing GFP or GFP-ctail in the auditory cortex. p=0.662. n=5 and 6 animals for GFP and c-tail, respectively. b. In vivo CALI with Z9139-eosin did not erase hippocampus-dependent IA fear memory. Latency period for re-entering the dark box of IA-trained mice with or without in vivo CALI. p=0.710. n=9 and 11 animals for CALI- and CALI+, respectively. Data were analyzed by Mann-Whitney U test. n.s. indicates no significance.

Supplementary Figure 11 Full-length of western blot by Z9139, anti-glua2, anti-glua3 and anti-actin antibody Full length immunoblots shown in Figure 1e. In GluA3 blots, asterisk indicates three lanes which was used in Figure 1e.

Supplementary Figure 12 Full-length of western blot by anti-arc and anti-gapdh antibodies. Full length immunoblots shown in Figure 4g. In GAPDH blots, asterisk indicates two lanes which was used in Figure 4g.