Volume 87, Issue 2, Pages (July 2015)

Slides:



Advertisements
Similar presentations
FMRP suppresses dendritic Arc protein levels.
Advertisements

Volume 70, Issue 4, Pages (May 2011)
Figure 1. Spine synapse density in LgDel mice is lower than in WT mice throughout postnatal development. (A) 3D reconstructed dendritic segments of WT.
Volume 77, Issue 2, Pages (January 2013)
Motor Protein KIF1A Is Essential for Hippocampal Synaptogenesis and Learning Enhancement in an Enriched Environment  Makoto Kondo, Yosuke Takei, Nobutaka.
Volume 7, Issue 3, Pages (May 2014)
Volume 112, Issue 3, Pages (February 2003)
Volume 18, Issue 3, Pages (September 2013)
Volume 9, Issue 4, Pages (November 2014)
Volume 82, Issue 1, Pages (April 2014)
Volume 54, Issue 6, Pages (June 2007)
Volume 88, Issue 3, Pages (November 2015)
Volume 14, Issue 6, Pages (June 2008)
Volume 56, Issue 1, Pages (October 2007)
Volume 87, Issue 2, Pages (July 2015)
Volume 78, Issue 1, Pages (April 2013)
Volume 55, Issue 5, Pages (September 2007)
Histamine Contributes to Tissue Remodeling via Periostin Expression
Volume 74, Issue 1, Pages (April 2012)
Activation of the Innate Signaling Molecule MAVS by Bunyavirus Infection Upregulates the Adaptor Protein SARM1, Leading to Neuronal Death  Piyali Mukherjee,
Volume 56, Issue 4, Pages (November 2007)
Volume 83, Issue 2, Pages (July 2014)
Volume 83, Issue 5, Pages (September 2014)
Volume 11, Issue 9, Pages (June 2015)
Volume 11, Issue 12, Pages (June 2015)
Volume 95, Issue 2, Pages e6 (July 2017)
The Temporal Dynamics of Arc Expression Regulate Cognitive Flexibility
Volume 77, Issue 2, Pages (January 2013)
Volume 78, Issue 4, Pages (May 2013)
A Role for Stargazin in Experience-Dependent Plasticity
Volume 76, Issue 2, Pages (October 2012)
Volume 91, Issue 2, Pages (July 2016)
Jungmook Lyu, Vicky Yamamoto, Wange Lu  Developmental Cell 
μ-Opioid Receptor and CREB Activation Are Required for Nicotine Reward
Volume 11, Issue 5, Pages (May 2015)
Volume 18, Issue 11, Pages (March 2017)
Volume 126, Issue 2, Pages (July 2006)
Experience-Dependent Equilibration of AMPAR-Mediated Synaptic Transmission during the Critical Period  Kyung-Seok Han, Samuel F. Cooke, Weifeng Xu  Cell.
Volume 20, Issue 13, Pages (September 2017)
Volume 57, Issue 2, Pages (January 2008)
Volume 23, Issue 3, Pages (April 2018)
Kentaro Abe, Masatoshi Takeichi  Neuron 
Rui Qin, Shuai Cao, Tianjie Lyu, Cai Qi, Weiguang Zhang, Yun Wang 
GRM7 Regulates Embryonic Neurogenesis via CREB and YAP
Bo Li, Ran-Sook Woo, Lin Mei, Roberto Malinow  Neuron 
Volume 65, Issue 3, Pages (February 2010)
Xuepei Lei, Jianwei Jiao  Stem Cell Reports 
PKA-GluA1 Coupling via AKAP5 Controls AMPA Receptor Phosphorylation and Cell- Surface Targeting during Bidirectional Homeostatic Plasticity  Graham H.
Volume 12, Issue 4, Pages (April 2007)
Matthew E. Klein, Pablo E. Castillo, Bryen A. Jordan  Cell Reports 
Volume 53, Issue 5, Pages (March 2007)
Gordon X. Wang, Stephen J Smith, Philippe Mourrain  Neuron 
Activin Signals through SMAD2/3 to Increase Photoreceptor Precursor Yield during Embryonic Stem Cell Differentiation  Amy Q. Lu, Evgenya Y. Popova, Colin.
Urtzi Garaigorta, Francis V. Chisari  Cell Host & Microbe 
Attenuated Cold Sensitivity in TRPM8 Null Mice
EVA1A/TMEM166 Regulates Embryonic Neurogenesis by Autophagy
Volume 16, Issue 11, Pages (September 2016)
Volume 18, Issue 12, Pages (March 2017)
Volume 151, Issue 7, Pages (December 2012)
Volume 17, Issue 20, Pages (October 2007)
Volume 51, Issue 4, Pages (August 2006)
Takashi Hayashi, Gavin Rumbaugh, Richard L. Huganir  Neuron 
Volume 90, Issue 3, Pages (May 2016)
Volume 71, Issue 6, Pages (September 2011)
FMRP Acts as a Key Messenger for Dopamine Modulation in the Forebrain
Volume 71, Issue 1, Pages (July 2011)
Arc/Arg3.1 Mediates Homeostatic Synaptic Scaling of AMPA Receptors
Arati Sridharan, Chetan Patel, Jit Muthuswamy 
Repulsive Guidance Molecule-a Is Involved in Th17-Cell-Induced Neurodegeneration in Autoimmune Encephalomyelitis  Shogo Tanabe, Toshihide Yamashita  Cell.
The Temporal Dynamics of Arc Expression Regulate Cognitive Flexibility
Presentation transcript:

Volume 87, Issue 2, Pages 382-398 (July 2015) Dysregulated ADAM10-Mediated Processing of APP during a Critical Time Window Leads to Synaptic Deficits in Fragile X Syndrome  Emanuela Pasciuto, Tariq Ahmed, Tina Wahle, Fabrizio Gardoni, Laura D’Andrea, Laura Pacini, Sébastien Jacquemont, Flora Tassone, Detlef Balschun, Carlos G. Dotti, Zsuzsanna Callaerts-Vegh, Rudi D’Hooge, Ulrike C. Müller, Monica Di Luca, Bart De Strooper, Claudia Bagni  Neuron  Volume 87, Issue 2, Pages 382-398 (July 2015) DOI: 10.1016/j.neuron.2015.06.032 Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 1 FMRP Regulates APP Protein Level during Postnatal Development (A) In Fmr1 KO mice, APP protein levels are upregulated during brain development. The protein expression was analyzed during development by WB in WT and KO mouse brain. For APP antibody specificity see Figures S2, S4, and S6B. Representative western blot showing protein levels at P7, P14, P21, P30, and P90. The histograms show quantified protein levels normalized to glyceraldehyde 3-phosphate dehydrogenase (GAPDH). This ratio was set to 1 in the WT mice for each developmental stage. The bars represent the SEM (∗p < 0.05; ∗∗p < 0.01, Student’s t test) (n = 4 for each developmental stage). (B) APP levels are increased in Fmr1 KO synaptoneurosomes. Representative western blot showing protein levels at P7, P14, P21, P30, and P90 in WT and Fmr1 KO mice. The histograms show quantified APP expression normalized to GAPDH. This ratio in WT mice was set to 1. The bars represent the SEM (∗p < 0.05; ∗∗p < 0.01, ∗∗∗p < 0.001, Student’s t test) (n = 3–4). See also Figure S1. Neuron 2015 87, 382-398DOI: (10.1016/j.neuron.2015.06.032) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 2 Impaired Processing of APP in Fmr1 KO Mice Leads to Enhanced sAPPα Release (A) Aβ40 levels in juvenile and aged Fmr1 KO cortices. The amount of Aβ was measured by ELISA normalized to the brain’s weight and expressed as the percentage of the WT. The bars represent the SEM (∗p < 0.05, Student’s t test) (n = 4 for both P21 and P300). (B) sAPP levels in KO brain cortices during postnatal development. The histograms show the quantification of sAPP protein levels normalized to Coomassie. The bars represent the SEM (∗p < 0.05, ∗∗p < 0.01, Student’s t test) (n = 3). (C) APP processing in P21 KO brain cortices. Representative WB showing protein levels of sAPP (sAPPα + sAPPβ), sAPPα, and sAPPβ in the soluble fraction (see also Figure S2) in WT and KO cortices. The histograms show the quantification of the three products normalized to Coomassie. The bars represent the SEM (∗p < 0.05, one-sample t test) (n = 4). (D) α- and β-secretase products in WT and Fmr1 KO synaptic membranes. APP, APP CTFα (∼10 kDa) and CTFβ (∼12 kDa), FMRP, and Vinculin protein levels were analyzed by WB in the synaptic membranes of WT and KO mice. The histograms show quantified proteins normalized to Vinculin. This ratio in WT mice was set to 1. The bars represent the SEM (∗p < 0.05, ∗∗p < 0.01, one-sample t test) (n = 6). See also Figure S2. (E) Cell-surface APP is reduced in Fmr1 KO cortical neurons. The WT and KO cortical neurons were stained for total APP (left) or surface APP (right). The representative dendritic fragments (>50 μm distance from the cell body) are shown. The histograms show the total and surface protein levels as the mean fluorescence intensity (Mean F.I.). The bars represent SEM (∗∗∗p < 0.001, Student’s t test) (n = 8 cells, five dendritic fragments [20 μm/cell]). The scale bar corresponds to 5 μm. (F) Surface APP is decreased in the Fmr1 KO cortical neurons. Proteins were biotinylated and captured with streptavidin-beads (input 1/10, lanes 1, 2 and 5; surface “sur” lanes 3, 4, and 6). APP, GAPDH, NR2A, and FMRP levels were analyzed by WB. The histograms show the quantification. The changes of total versus surface APP are expressed as ratio of the WT. The bars represent the SEM (∗p < 0.05, ∗∗p < 0.01, one-sample t test) (n = 4). (G) sAPP levels are increased in cortical neurons. sAPP levels were analyzed by WB in the conditioned medium from WT and KO cortical neurons. The histograms show the quantification of APP protein levels normalized to the Coomassie. This ratio was set to 1 in the WT mice. The bars represent the SEM (∗p < 0.05, Student’s t test) (n = 4). Neuron 2015 87, 382-398DOI: (10.1016/j.neuron.2015.06.032) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 3 FMRP Regulates the APP Processing Enzyme ADAM10 (A) ADAM10 expression is higher in Fmr1 KO mice than in WT. Representative WB showing the ADAM10 protein levels in WT and KO cortices (black arrowheads: mature proteins and white arrowheads: prodomain containing proteins). The histograms show the quantification of ADAM10 and APP protein levels normalized to GAPDH levels expressed as ratio of the WT. The bars represent the SEM (∗p < 0.05, ∗∗p < 0.01, Student’s t test) (n = 3). (B) ADAM10 expression is increased in synaptoneurosomes (Syn) from KO mice. Representative WB showing ADAM10, ADAM9, and ADAM17 expression levels in the WT and the KO mice (black arrowheads: mature protein and white arrowhead: immature protein). The histograms represent the quantification of protein levels normalized to GAPDH levels expressed as ratio of the WT. The bars represent the SEM (∗∗p < 0.01, Student’s t test) (n = 4). (C) ADAM10 expression during development. ADAM10 protein levels were detected in WT and KO brain cortex extracts at P14, P21, P30, and P90. The GAPDH was used as normalizer. The bars represent the SEM ( ∗∗p < 0.01, Student’s t test) (n = 3). (D) Immunofluorescence of ADAM10 in DIV14 cortical neurons. The left image shows representative dendritic fragments (>50 μm from cell body). The histograms represent protein levels quantified as the mean fluorescence intensity (Mean F.I.). The bars represent SEM (∗∗∗p < 0.001, Student’s t test) (n = 10 cells, five denditic fragments [20 μm/cell]). The scale bar represents 5 μm. (E) Surface ADAM10 is increased in Fmr1 KO cortical neurons. Proteins were biotinylated and captured with streptavidin-Dynabeads; ADAM10 protein levels were analyzed by WB. The histograms show the quantification of the protein levels. The bars represent the SEM (∗p < 0.05, ∗∗p < 0.01, one-sample t test) (n = 4). (F) Detection of FMRP-associated mRNAs. The top panel shows a representative WB of FMRP-IP from brain extracts. Panels below show (RT-PCR to detect App, Adam10, Adam9, and Adam17 mRNAs). Negative controls: dopamine receptor D2 (D2DR) and (Cyp46) mRNAs. Positive control: αCaMKII mRNA. Lane 1, input (1/20); lane 2, αFMRP; and lane 3, control IgGs. (G) Translational efficiency of App and Adam10 mRNAs in WT and Fmr1 KO cortices. The extracts were fractionated along a 10%–60% sucrose gradient. There were 12 fractions that were collected while reading the absorbance at 254 nm. The amount of Adam10, App and β-actin mRNAs in each fraction was quantified by RT-quantitative (q) PCR. A representative polysomal-mRNP profile is shown. Each fraction shows the quantification of App and Adam10 mRNAs normalized to β-actin mRNA in the WT and Fmr1 KO mice. The bars represent the SEM (∗p < 0.05, ∗∗p < 0.01, Student’s t test) (n = 4). Neuron 2015 87, 382-398DOI: (10.1016/j.neuron.2015.06.032) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 4 sAPPα Levels Are Crucial for Spine Morphology in FXS (A) Representative dendritic segments of cultured cortical neurons transfected with lentiviral vectors expressing EGFP-App shRNA or EGFP-CTRL shRNA, treated with sAPPα at DIV8, and analyzed at DIV14. The scale bar represents 5 μm. (B) Spine density of the different spine types. The bars represent the SEM (∗∗∗p < 0.001, non-significant = n.s., one-way ANOVA followed by post hoc Bonferroni correction) (n = 10 neurons from at least two different cultures). For each condition, 350–500 spines were analyzed. (C) Density of the mature and immature spines. The spine morphology was used to discriminate the spine types. See Figure S5. The bars represent the SEM (∗∗∗p < 0.001, one-way ANOVA followed by post hoc Bonferroni correction) (n = 10 neurons from at least two different cultures). For each condition, 350–500 spines were analyzed. See also Figures S4 and S5. Neuron 2015 87, 382-398DOI: (10.1016/j.neuron.2015.06.032) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 5 Exaggerated Protein Synthesis in Fmr1 KO Depends on Excessive ADAM10 Activity and sAPPα Production (A) Genetic reduction of APP in Fmr1 KO mice ameliorates the excessive protein synthesis in cortical neurons. Representative WB showing incorporated puromycin in synaptoneurosomes. The histograms show the quantification of puromycin incorporation normalized to Coomassie. The bars represent the SEM (∗p < 0.05 KO versus WT, # p < 0.05 KO versus Fmr1 KO/AppHet, one-sample t test) (Holm’s correction) (n = 4 independent cultures). (B) Protein synthesis in Fmr1 KO/AppHet mice in synaptoneurosomes. Representative WB showing incorporated puromycin. The histogram shows a quantification of puromycin incorporation normalized to GAPDH. The bars represent the SEM. Kruskal-Wallis test followed by Dunn’s post hoc test. (∗p < 0.05) (n = 4 WT; n = 4 Fmr1 KO; and n = 3 Fmr1 KO/AppHet). (C) sAPPα induces de novo protein synthesis. WT cortical neurons were treated with 1 nM sAPPα for 1 hr and analyzed as above. The bars represent the SEM (∗p < 0.05, one-sample t test) (n = 3). (D) sAPPα increases translation in the Fmr1 KO/AppHet. Cortical neurons were treated with 1 nM sAPPα for 1 hr. The puromycin uptake was quantified by WB. The bars represent the SEM (∗p < 0.05, one-sample t test) (n = 3). (E) Basal protein synthesis in Fmr1 KO/Adam10Het synaptoneurosomes. Representative WB showing incorporated puromycin in synaptoneurosomes. The histograms show a quantification of puromycin incorporation normalized to GAPDH. The bars represent the SEM. Kruskal-Wallis test followed by Dunn’s post hoc test. (∗p < 0.05, ### p < 0.001 KO versus Fmr1 KO/Adam10Het) (n = 4 WT; n = 4 Fmr1 KO; and n = 3 Fmr1 KO/Adam10Het). (F) sAPPα promotes mGluR1/5 signaling. Cortical neurons were treated for 1 hr with sAPPα with or without MPEP 50 μM or with Rapamycin 200 nM. Representative WB showing ERK 1/2 and mTOR phosphorylation levels. The histograms show a quantification of the protein levels normalized to Vinculin. The bars represent the SEM (∗p < 0.05, one-way ANOVA followed by Dunnett’s multiple comparisons test) (n = 5 independent cultures). Neuron 2015 87, 382-398DOI: (10.1016/j.neuron.2015.06.032) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 6 Modulation of ADAM10 Activity Reduces Excessive sAPPα and Ameliorates LTD and Behavior in the Fmr1 KO Mice (A) Left scheme: TAT-Pro (in black) perturbs the ADAM10/SAP97 association and impairs ADAM10 (A10) localization at the cell surface. The treatment with TAT-Pro reduces sAPP release in neurons. The DIV15 cortical neurons were treated with TAT-Ala and TAT-Pro peptides (10 μM, 18 hr). The media was collected and sAPP levels were measured by WB (middle). The histograms show the quantification of sAPP normalized to Coomassie staining. The bars represent the SEM (∗p < 0.05, ∗∗p < 0.01, versus TAT-Ala WT; # p < 0.05, TAT-Pro KO versus TAT-Ala KO, one-way ANOVA and Sidak’s multiple comparisons test) (n = 3). (B) ADAM10 activity can be modulated in vivo in the Fmr1 KO. The juvenile mice (P21) received a single intraperitoneal injection of either a TAT-Pro (3 nmol/g) or TAT-Ala peptide (3 nmol/g). The effect on sAPPα release in the WT and KO brain cortices was monitored by WB after 18 hr. The histograms show the quantification of sAPPα protein levels normalized to Coomassie. The bars represent the SEM (∗p < 0.05, TAT-Ala WT versus TAT-Ala KO; ## p < 0.01, TAT-Pro KO versus TAT-Ala KO, one-way ANOVA and Sidak’s multiple comparisons test) (n = 5). (C) Fmr1 KO mice exhibit enhanced mGluR-LTD (n = 15 slices from nine mice/genotype). (D) The mice (P18-P25) received a single intraperitoneal injection of either a Tat-Pro (2 nmol/g) or TAT-Ala peptide (2 nmol/g). TAT-Ala or TAT-Pro peptide does not impact LTD in the WT mice, whereas (E) TAT-Pro prevents mGluR-LTD in the Fmr1KO mice (F = 20, 75, p < 0.0002) (TAT-Pro n = 9 slices from six mice and TAT-Ala n = 8 slices from four mice). Solid bars indicate the duration of the bath application of DHPG (30 μM, 15 min). The representative traces (right) showing fEPSP before (1), 10 min after (2), and 240 min after (3) DHPG application. The stimulus artifact is blanked to ease interpretation. The data are shown as the means ± SEM. See also Figure S8. (F) In vivo effects of TAT-Pro peptide treatment on protein expression in cortex. APP, ADAM10, ARC, and STEP protein levels were analyzed by WB. The bars represent the SEM (∗p < 0.05, versus WT; #p < 0.05, TAT-Pro Ko versus KO, one-way ANOVA and Sidak’s multiple comparisons test) (n = 5). (G) Effect of the TAT-Pro peptide treatment on the performance of WT and Fmr1 KO mice in the open field. Juvenile mice (P21) have been tested after 2 consecutive days of treatment with the peptide (2 nmol/g). Histograms represent the distance and the speed of the animals in the open field. (one-way ANOVA followed by Sidak’s multiple comparisons test, and ∗p < 0.05). (n = 15 WT; n = 11 Fmr1 KO; n = 14 TAT-Pro WT; and n = 17 TAT-Pro Fmr1 KO). (H) Effect of the peptide in the T-maze. The table shows the preference index for the novel arm in test 2 (1, no preference; >1, preference for the Novel Arm; and <1 preference for Familiar Arm. The histograms show the preference for the novel arm in the test 2 of the T-maze test (∗∗∗p < 0.001, Chi square test) (n = 9 WT; n = 9 Fmr1 KO; n = 9 TAT-Pro WT; and n = 12 TAT-Pro Fmr1 KO). (I) Effect of the TAT-Pro peptide treatment on the nest building performance of WT and Fmr1 KO after 48 hr. The left histogram show the measure of the unused cotton cylinders (one-way ANOVA followed by Sidak’s multiple comparisons test, ∗p < 0.05, and ∗∗∗p < 0.001) (n = 8 WT; n = 8 Fmr1 KO; n = 5 TAT-Pro WT; and n = 8 TAT-Pro Fmr1 KO). The right images show representative examples of nest building in the tested conditions. Neuron 2015 87, 382-398DOI: (10.1016/j.neuron.2015.06.032) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 7 APP and ADAM10 Expression in Fragile X Patients (A) APP and ADAM10 protein levels in human lymphoblastoid cells. Representative WB of APP, ADAM10, and FMRP protein levels in cell lines from healthy controls and FXS. The images belong to the same WB. The histograms show the quantification of APP, ADAM10, and FMRP normalized to Vinculin. The bars represent the SEM (∗p < 0.05, ∗∗p < 0.01, and Student’s t test) (n = 6 controls, 6 FXS). (B) APP and ADAM10 levels in the cortex of FXS patients. The upper images show representative WB of APP and ADAM10 levels in the cortex of healthy controls and FXS (Table S1). The histograms show the quantification of APP, ADAM10, and FMRP normalized to Vinculin. The bars represent the SEM ( ∗p < 0.05, ∗∗p < 0.01, Student’s t test the prefrontal cortex) (n = 8 controls, 7 FXS). (C) APP and ADAM10 levels in human fibroblasts. Shown are representative WBs detecting APP and ADAM10 levels in cells from healthy controls and FXS patients (left), and the quantification of APP (middle) and ADAM10 (right), normalized to GAPDH. The bars represent the SEM (∗p < 0.05, Mann-Whitney test) (n = 7 controls, 8 FXS, at least three independent experiments shown in the graph). (D) sAPPα in human FXS fibroblasts. Representative WB shows the levels of sAPPα in the media of control and FXS cells. (E) IP of FMRP-associated mRNAs in human fibroblasts. RT-qPCR was used to detect APP and ADAM10 mRNAs. The histograms represent the level of FMRP-bound mRNAs, the subtracted background was obtained by the IgGs. The average of the negative controls (CTRL): Vinculin (VCL), β-Glucuronidase (GUSB), and Hypoxanthineribosyltransferase (HPRT) mRNAs was set at 1 in each experiment (n = 3 controls, two independent experiments). The bars represent the SEM (∗p < 0.05, one sample t test). Neuron 2015 87, 382-398DOI: (10.1016/j.neuron.2015.06.032) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 8 Restoring Synaptic Homeostasis in FXS Left panel: in physiological conditions (WT) FMRP regulates the expression of several synaptic proteins downstream the mGluR1/5 signaling pathway. Among them it regulates the expression of APP and the α-secretase ADAM10. APP is cleaved by ADAM10 to generate sAPPα. sAPPα signals through the mGluR1/5 and activates the MAPK pathway, ultimately regulating protein translation affecting proper spine morphology, synaptic plasticity, and behavior. The right image shows that loss of FMRP (FXS) compromises the fine-tuned expression of a variety of synaptic proteins. This lack of translational control results in combined upregulation of APP and ADAM10 and accumulation of sAPPα. sAPPα triggers ERK1/2 phosphorylation and sustains mGluR1/5 activation and protein translation. Dysregulation of the APP-ADAM10 processing pathway leads to FXS deficits. Finally, reduction of ADAM10 trafficking/activity at synapses, using the TAT-Pro peptide, restores sAPPα levels, mGluR-LTD, protein expression, and behavioral deficits in FXS mice. Neuron 2015 87, 382-398DOI: (10.1016/j.neuron.2015.06.032) Copyright © 2015 Elsevier Inc. Terms and Conditions