+ All Categories
Home > Documents > Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia,...

Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia,...

Date post: 01-Aug-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
10
SHORT REPORT Regulation of dendrite morphology and excitatory synapse formation by zDHHC15 Bhavin S. Shah*, Jordan J. Shimell* and Shernaz X. Bamji ABSTRACT Protein palmitoylation is the most common post-translational lipid modification in the brain and is mediated by a family of 24 zDHHC enzymes. There has been growing interest in zDHHCs due to mounting evidence that these enzymes play key roles in the development and function of neuronal connections, and the fact that a number of zDHHCs have been associated with neurodevelopmental and neurodegenerative diseases. Loss-of-function variants in several zDHHCs, including zDHHC15, have been identified in patients with intellectual disabilities; however, the function of zDHHC15 in the brain has not been well studied. Here, we demonstrate that knocking down zDHHC15 in primary rat hippocampal cultures reduces dendritic outgrowth and arborization, as well as spine maturation. Moreover, knockdown of zDHHC15 reduces palmitoylation of PSD-95 and its trafficking into dendrites, resulting in an overall decrease in the density of excitatory synapses being formed onto mutant cells. KEY WORDS: zDHHC15, Palmitoyl transferases, Palmitoylation, Excitatory synapses, PSD-95, DLG4, Dendrite morphology INTRODUCTION Post-translational modification of cellular proteins by S-acylation involves the reversible attachment of fatty acids to cysteine residues, and is important for the trafficking of proteins towards cell membranes and the regulation of cellular signaling (Fukata and Fukata, 2010; Ko and Dixon, 2018). Palmitoylation is the reversible attachment of the fatty acid palmitic acid, and is the most common post-translational lipid modification in the brain (Fukata and Fukata, 2010). Enzymes that mediate palmitoylation consist of a family of 23 proteins containing a conserved Asp-His-His-Cys (DHHC) motif that is required for enzymatic activity (Fukata and Fukata, 2010). Nine of the 23 genes encoding zinc finger DHHC-type (zDHHC) enzymes have been associated with brain disorders and 41% of all identified synaptic proteins are substrates for palmitoylation (Sanders et al., 2015), underscoring the importance of palmitoylation in the development and function of the brain and, specifically, in synapse biology. Despite their implied importance, little is known about the role of zDHHC enzymes in the brain and whether disrupting their enzymatic activity contributes to brain pathology (Charollais and Van Der Goot, 2009; Fukata and Fukata, 2010; Rocks et al., 2010). Although zDHHC15 is highly expressed in many cell types in the brain (Fukata et al., 2004; Mansouri et al., 2005; Wang et al., 2015; Zhang et al., 2014), little is known about its function. Known substrates of zDHHC15 include PSD-95 (officially known as DLG4), GAP43, SNAP25b, CSP (officially known as DNAJC5), GABARγ2 (officially known as GABRG2) , Fyn, BACE1, CD151, CIMPR (officially known as IGF2R) and SORT1 (Fang et al., 2006; Fukata et al., 2004; Greaves et al., 2010, 2008; Mill et al., 2009; Sharma et al., 2008; Tsutsumi et al., 2009; Vetrivel et al., 2009; Yokoi et al., 2016), proteins that have been shown to play an important role in the development and function of neuronal connections. ZDHHC15 is one of a number of genes on the X chromosome that is duplicated in patients with intellectual disability (Linhares et al., 2016; Martinez et al., 2014), and one report has identified the loss of a ZDHHC15 transcript in a female patient with non-syndromic X-linked intellectual disability (Mansouri et al., 2005). Our study characterizes the role of zDHHC15 in the development of neuronal connectivity. zDHHC15 is highly expressed at embryonic stages and reduced in adult brains. Knockdown of zDHHC15 in hippocampal cultures reduces dendritic arborization and spine maturation. Notably, zDHHC15 knockdown results in a marked reduction in the density of excitatory synapses, resulting from reduced palmitoylation of PSD-95 and its trafficking into dendrites. RESULTS AND DISCUSSION zDHHC15 is expressed during early stages of brain development, and in cultured excitatory and inhibitory hippocampal neurons ZDHHC15 mRNA is abundantly expressed in the brain compared to other tissues (Fukata et al., 2004). After validating the specificity of our zDHHC15 antibody (Fig. S1A-C), we determined that zDHHC15 is highly expressed during early stages of development i.e. at embryonic day 17 (E17) and postnatal day 10 (P10) and significantly reduced in the adult (P90) brain (Fig. 1A,B). To further examine the cellular and subcellular distribution of zDHHC15, we immunostained primary hippocampal cultures at 13 days in vitro (DIV). zDHHC15 was observed in all neurons (identified by using anti-MAP2 antibody) including GAD65- positive inhibitory neurons, demonstrating that zDHHC15 is expressed in both excitatory and inhibitory neurons. In contrast, in these young cultures, zDHHC15 was not expressed in GFAP- expressing glial cells (Fig. 1C). zDHHC enzymes have been shown to localize to the plasma membrane and/or organelle membranes (Ohno et al., 2006). To determine whether zDHHC15 is localized to the plasma membrane, we performed a surface biotinylation assay using neurons at 13 DIV (Fig. 1D). Unlike zDHHC5 or zDHHC8, which have previously been shown to localize to plasma membranes (Brigidi et al., 2015; Ohno et al., 2006), zDHHC15 was not isolated in surface fractions, Received 23 January 2019; Accepted 28 May 2019 Department of Cellular & Physiological Sciences & the Brain Research Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed equally to this work Author for correspondence ([email protected]) B.S.S., 0000-0001-6594-5871; S.X.B., 0000-0003-0102-9297 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 1 © 2019. Published by The Company of Biologists Ltd | Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052 Journal of Cell Science
Transcript
Page 1: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

SHORT REPORT

Regulation of dendrite morphology and excitatory synapseformation by zDHHC15Bhavin S. Shah*, Jordan J. Shimell* and Shernaz X. Bamji‡

ABSTRACTProtein palmitoylation is the most common post-translational lipidmodification in the brain and is mediated by a family of 24 zDHHCenzymes. There has been growing interest in zDHHCs due tomounting evidence that these enzymes play key roles in thedevelopment and function of neuronal connections, and the fact thata number of zDHHCs have been associated with neurodevelopmentaland neurodegenerative diseases. Loss-of-function variants in severalzDHHCs, including zDHHC15, have been identified in patients withintellectual disabilities; however, the function of zDHHC15 in the brainhas not been well studied. Here, we demonstrate that knocking downzDHHC15 in primary rat hippocampal cultures reduces dendriticoutgrowth and arborization, as well as spine maturation. Moreover,knockdown of zDHHC15 reduces palmitoylation of PSD-95 and itstrafficking into dendrites, resulting in an overall decrease in the densityof excitatory synapses being formed onto mutant cells.

KEY WORDS: zDHHC15, Palmitoyl transferases, Palmitoylation,Excitatory synapses, PSD-95, DLG4, Dendrite morphology

INTRODUCTIONPost-translational modification of cellular proteins by S-acylationinvolves the reversible attachment of fatty acids to cysteine residues,and is important for the trafficking of proteins towards cellmembranes and the regulation of cellular signaling (Fukata andFukata, 2010; Ko and Dixon, 2018). Palmitoylation is the reversibleattachment of the fatty acid palmitic acid, and is the most commonpost-translational lipid modification in the brain (Fukata and Fukata,2010). Enzymes that mediate palmitoylation consist of a family of23 proteins containing a conserved Asp-His-His-Cys (DHHC)motif that is required for enzymatic activity (Fukata and Fukata,2010). Nine of the 23 genes encoding zinc finger DHHC-type(zDHHC) enzymes have been associated with brain disordersand ∼41% of all identified synaptic proteins are substrates forpalmitoylation (Sanders et al., 2015), underscoring the importanceof palmitoylation in the development and function of the brain and,specifically, in synapse biology. Despite their implied importance,little is known about the role of zDHHC enzymes in the brain andwhether disrupting their enzymatic activity contributes to brain

pathology (Charollais and Van Der Goot, 2009; Fukata and Fukata,2010; Rocks et al., 2010).

Although zDHHC15 is highly expressed in many cell types in thebrain (Fukata et al., 2004; Mansouri et al., 2005; Wang et al., 2015;Zhang et al., 2014), little is known about its function. Knownsubstrates of zDHHC15 include PSD-95 (officially known asDLG4),GAP43, SNAP25b, CSP (officially known as DNAJC5), GABARγ2(officially known as GABRG2) , Fyn, BACE1, CD151, CIMPR(officially known as IGF2R) and SORT1 (Fang et al., 2006; Fukataet al., 2004; Greaves et al., 2010, 2008; Mill et al., 2009; Sharmaet al., 2008; Tsutsumi et al., 2009; Vetrivel et al., 2009; Yokoi et al.,2016), proteins that have been shown to play an important role in thedevelopment and function of neuronal connections.ZDHHC15 is oneof a number of genes on the X chromosome that is duplicated inpatients with intellectual disability (Linhares et al., 2016; Martinezet al., 2014), and one report has identified the loss of a ZDHHC15transcript in a female patient with non-syndromic X-linkedintellectual disability (Mansouri et al., 2005).

Our study characterizes the role of zDHHC15 in the developmentof neuronal connectivity. zDHHC15 is highly expressed at embryonicstages and reduced in adult brains. Knockdown of zDHHC15 inhippocampal cultures reduces dendritic arborization and spinematuration. Notably, zDHHC15 knockdown results in a markedreduction in the density of excitatory synapses, resulting from reducedpalmitoylation of PSD-95 and its trafficking into dendrites.

RESULTS AND DISCUSSIONzDHHC15 is expressed during early stages of braindevelopment, and in cultured excitatory and inhibitoryhippocampal neuronsZDHHC15mRNA is abundantly expressed in the brain compared toother tissues (Fukata et al., 2004). After validating the specificity ofour zDHHC15 antibody (Fig. S1A-C), we determined that zDHHC15is highly expressed during early stages of development – i.e. atembryonic day 17 (E17) andpostnatal day 10 (P10) – and significantlyreduced in the adult (P90) brain (Fig. 1A,B).

To further examine the cellular and subcellular distribution ofzDHHC15, we immunostained primary hippocampal cultures at13 days in vitro (DIV). zDHHC15 was observed in all neurons(identified by using anti-MAP2 antibody) – including GAD65-positive inhibitory neurons, demonstrating that zDHHC15 isexpressed in both excitatory and inhibitory neurons. In contrast, inthese young cultures, zDHHC15 was not expressed in GFAP-expressing glial cells (Fig. 1C).

zDHHC enzymes have been shown to localize to the plasmamembrane and/or organelle membranes (Ohno et al., 2006). Todetermine whether zDHHC15 is localized to the plasma membrane,we performed a surface biotinylation assay using neurons at 13 DIV(Fig. 1D). Unlike zDHHC5 or zDHHC8, which have previouslybeen shown to localize to plasma membranes (Brigidi et al., 2015;Ohno et al., 2006), zDHHC15 was not isolated in surface fractions,Received 23 January 2019; Accepted 28 May 2019

Department of Cellular & Physiological Sciences & the Brain Research Centre,University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C.,V6T-1Z3, Canada.*These authors contributed equally to this work

‡Author for correspondence ([email protected])

B.S.S., 0000-0001-6594-5871; S.X.B., 0000-0003-0102-9297

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

1

© 2019. Published by The Company of Biologists Ltd | Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 2: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

Fig. 1. zDHHC15 is expressed during early stages of neocortical development and in Golgi compartments of cultured excitatory and inhibitoryneurons. (A,B) Western blot and quantification of zDHHC15 protein levels in the hippocampus (Hc), cerebral cortex (C), hindbrain (Hb) or cerebellum (Cb) ofmouse brain at E17, P10 and P90. Levels of zDHHC15 decreased from E17 to P90 in these regions of mouse brain. n=4 tissue samples per time point,*P<0.05, **P<0.001, one-way ANOVA with Tukey’s post-hoc test, means±s.e.m. (C) Representative confocal images of 13 DIV rat hippocampal cultures. Allneurons, identified by using anti-MAP2 antibody (white arrows), expressed zDHHC15 (yellow arrows) – including inhibitory GAD65-positive neurons(white arrowheads). GFAP-positive glial cells (green arrows) exhibited weak to no zDHHC15 immunostaining. White arrows denote DAPI-stained nuclei thatare positive for zDHHC15, whereas green arrows denote DAPI-stained nuclei that are negative for zDHHC15. (D) Western blot of lysates from biotinylatedneurons at 13 DIV probed for zDHHC5, zDHHC8 and zDHHC15; the latter is not localized at the membrane. n=3 cultures. (E) zDHHC15 colocalizeswith the cis-medial Golgi marker giantin. (F-H) Microimages (F) showing 13 DIV rat hippocampal neurons stained for zDHHC15 and the synaptic markersPSD-95 and gephyrin. zDHHC15 colocalizes with a small subset of PSD-95 or gephyrin puncta. zDHHC15 and PSD-95 images were masked as per Fig. S2E.n=10 neurons, three cultures. Scale bars: 100 µm (C), 20 µm (E, top) and 10 μm (E, bottom; F).

2

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 3: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

suggesting it is restricted to internal membranes. Coimmunostainingwith zDHHC15 and the Golgi marker giantin demonstrated thatzDHHC15 is highly expressed in the somatic Golgi complex as wellas in dendrites – with 67±3% of all zDHHC15-positive punctalocalizing with giantin (Fig. 1E,F). The localization of zDHHC15 inthe somatic Golgi of neurons is in accordance to a previous reportdemonstrating zDHHC15 localization to the Golgi compartment ofHEK293 cells (Greaves et al., 2008) and hippocampal neurons(Levy et al., 2011).As zDHHC15 was distributed in a punctate manner within

dendrites, we determined whether zDHHC15 localized to PSD-95and gephyrin puncta that – as demonstrated by us – are faithfulexcitatory and inhibitory markers, respectively (Fig. S2A-D). Thedensity of colocalized zDHHC15 and PSD-95, and zDHHC15 andgephyrin puncta was quantified within eGFP ‘masks’ of neurons(Fig. S2E). Coimmunostaining demonstrated minimal localizationof zDHHC15 at synapses (Fig. 1G). Only 18±2% of zDHHC15-positive puncta colocalized with PSD-95 and 10±2% colocalizedwith gephyrin (in turn 22±2% of PSD-95-positive and 23±3% ofgephyrin-positive puncta colocalized with zDHHC15) (Fig. 1G,H).Although the modest colocalization of PSD-95 and zDHHC15 mayappear surprising – as PSD-95 is a known substrate of zDHHC15(Fukata et al., 2004) – zDHHC15 function is thought to bedependent on synaptic activity (Noritake et al., 2009). It is,therefore, possible that the subcellular localization of zDHHC15 is

altered following changes in synaptic activity. Together, these datasuggest that zDHHC15 mediates its function during early stages ofneuronal development, and that its primary site of action is at Golgicompartments that are distinct from Golgi satellites associated withexcitatory or inhibitory synapses.

zDHHC15 promotes dendritic outgrowth and arborizationIn humans, the ZDHHC15 gene is located on the X-chromosome andloss of ZDHHC15 transcript has previously shown to be associatedwith intellectual disabilities (Mansouri et al., 2005). Based on this andthe high expression of zDHHC15 protein during development, wehypothesized that zDHHC15 is important for neuronal connectivity.To test this, we generated small hairpin RNA targeting zDHHC15(shRNA; validated in Fig. 2A) and examined whether zDHHC15knockdown impacts neuronal morphology (see Fig. S3A,B for rawimages, neuron masking, tracing and Sholl analysis). Neuronsexpressing zDHHC15 shRNA exhibited significantly shorterdendrites (Fig. 2B,C), decreased branch count (Fig. 2B,D) andless dendritic complexity (Fig. 2B,E) compared to neuronstransfected with control shRNA. These dendritic defects wererescued in cells expressing an shRNA-resistant zDHHC15 construct(zDHHC15R) (Fig. 2B-E). To determine whether the palmitoylationfunction of zDHHC15 is important for dendritic outgrowth, cellswere co-transfected with shRNA and palmitoylation-deficientzDHHC15, in which the cysteine residue of DHHC had been

Fig. 2. zDHHC15 promotes dendritic growth and arborization. (A) Western blot of rat hippocampal neurons nucleofected at 0 DIV, lysed at 4 DIV and probedwith indicated antibodies. n=3 cultures. (B) Rat hippocampal neurons were transfected with the indicated constructs, fixed and imaged at 13 DIV. Imagesshown have been manually masked to remove background axons; raw images are available in Fig. S3A with an example of masking, tracing and Sholl analysisavailable in Fig. S3B. Scale bar: 100 µm. (C-E) Quantification of dendrite length and counts from 13 DIV rat hippocampal neurons. Knockdown of zDHHC15(using shRNA targeting zDHHC15, referred to as shRNA) decreases dendritic length and complexity. Statistical significance was calculated for the entireSholl profile. n=43 (control), 49 (shRNA), 34 (shRNA+zDHHC15), 40 (shRNA+zDHHS15), 36 (zDHHC15) neurons, three cultures. **P<0.01, ***P<0.001, oneway ANOVA and Tukey’s post-hoc test; mean±s.e.m.

3

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 4: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

changed to serine (zDHHS15R). Although expression ofzDHHS15R was similar to that of zDHHC15R (Fig. 2A), it wasunable to rescue the knockdown phenotype, indicating that theenzymatic activity of zDHHC15 is essential for zDHHC15-mediated regulation of dendritic arbor length and complexity(Fig. 2B-E). Overexpression of zDHHC15 had no effect on dendritelength, number or complexity (Fig. 2B-E).To further investigate whether zDHHC15 promotes dendritic

outgrowth or stabilization, neurons were imaged every 24 h for 72 h

post transfection using time-lapse imaging. At 72 h posttransfection, cells expressing zDHHC15 shRNA exhibited anincrease of only 13±4.7% in total dendritic length, whereasdendrite length of cells expressing control shRNA,shRNA+zDHHC15R or zDHHC15 alone increased by 33±3.9%,34±4.0% or 39±6.1%, respectively (Fig. 3A,B, raw imagesFig. S3C). These data indicate that zDHHC15 is required fordendritic outgrowth and not stability, because knockdown ofzDHHC15 inhibited further dendritic outgrowth.

Fig. 3. zDHHC15 knockdown leads to inhibitionof dendritic outgrowth and formation of maturespines. (A) Representative, masked, time-lapseconfocal images of neurons transfected at 10 DIV witheGFP plus the indicated constructs and imaged 24, 48and 72 h post transfection. Raw images are availablein Fig. S3C. Scale bar: 100 µm. (B) zDHHC15knockdown inhibits dendrite growth. n=10 neurons,three cultures. (C-E) Hippocampal neurons at 10 DIVtransfected with the indicated constructs, fixed, andimaged at 13 DIV to calculate the density (C) andtype (D,E) of spiny protrusions. While there was nosignificant change in the density of overall protrusions(C), zDHHC15 knockdown significantly increased theproportion of filopodia (D) and reduced the proportionof mature, mushroom and stubby spines (E). n=56(control), 43 (shRNA), 38 (shRNA+zDHHC15),33 (shRNA+zDHHS15), 31 (zDHHC15) neurons, threecultures. ***P<0.001; ns, not significant; one-wayANOVA and Tukey’s post-hoc test (C-E), mean±s.e.m.

4

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 5: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

zDHHC15 promotes spine maturation and the formationof excitatory synapsesWe next determined whether zDHHC15 regulates the formation and/or maturation of spiny protrusions – including filopodia – that exhibitdynamic motility and are associated with smaller, less-maturesynapses, as well as with stubby or mushroom spines, which aretypically associated with large, mature synapses (Casanova et al.,2012; Marin-Padilla, 1972). Although knockdown of zDHHC15 didnot impact the total number of spiny protrusions at 13–14 DIV(Fig. 3C), it resulted in a significant increase in the proportion of thin,immature protrusions, and a decrease in the proportion of mature,stubby and mushroom spines (Fig. 3D,E). Although zDHHC15R

rescued this knockdown phenotype, palmitoylation-defectivezDHHS15R did not, indicating that zDHHC15 regulates spinemorphology and maturation through its palmitoylation function.As the excitatory postsynaptic protein PSD-95 can be

palmitoylated by zDHHC15 (as well as by zDHHC2, zDHHC3and zDHHC7) (Fukata et al., 2004), and because knockdown ofzDHHC15 decreases the proportion of mature spines (Fig. 3D,E), wenext investigated whether zDHHC15 regulates the formation and/ormaintenance of synaptic connections. At 10 DIV, cultures weretransfected with eGFP plus the constructs indicated in Fig. 4A, maskswere generated as previously described (Fig. S2E) (Brigidi et al.,2015), and cells immunostained for PSD-95 and gephyrin.zDHHC15 knockdown led to a significant reduction in the densityof puncta positive for PSD-95 but not gephyrin (Fig. 4A,B), causingan overall decrease in the ratio of excitatory to inhibitory (E:I)synapses (Fig. 4C). Of note, zDHHC15 knockdown also resulted in adecrease in the density of colocalized PSD-95 and/or VGlut1(vesicular glutamate transporter 1, an excitatory presynaptic marker)demonstrating a reduction in excitatory synapse number, and not justPSD-95 density (Fig. 4D, Fig. S2F). While expression of wild-typezDHHC15R rescued the knockdown phenotype, zDHHS15R did not,indicating that the enzymatic activity of zDHHC15 is important for itsregulation of excitatory synapse density (Fig. 4A-C).As PSD-95 is also palmitoylated by zDHHC2, zDHHC3 and

zDHHC7 (Fukata et al., 2004), we determinedwhether overexpressionof these enzymes can rescue the zDHHC15 knockdown phenotype(we did not use zDHHC7 as its levels are low in the hippocampus)(https://www.proteinatlas.org/ENSG00000153786-ZDHHC7/tissue)and proved difficult to express (validation of zDHHC2 and zDHHC3expression, Fig. S4). The zDHHC15 shRNA-mediated decrease inPSD-95 density was rescued by zDHHC3 and zDHHC15 and, to alesser extent, zDHHC2 (Fig. 4D). Moreover, while zDHHC15knockdown did not impact PSD-95 puncta size, coexpression ofzDHHC15 shRNA with zDHHC2 enhanced the area of PSD-95clusters by around 10%, suggesting a different mechanism of action(Fig. 4E). In concordance with zDHHC2 and zDHHC3’s ability torescue PSD-95 density, expression of these proteins rescued spinematuration (Fig. 4F,G). However, in contrast to the ability of zDHHC2and zDHHC3 to rescue synapse density and spine maturation, neitherzDHHC2 nor zDHHC3 rescued zDHHC15 knockdown-mediatedchanges regarding the dendritic length (Fig. 4H). These resultsdemonstrate that zDHHC15 knockdown decreases excitatory synapsedensity and outgrowth, and that the pathway regulating excitatorysynapse density is independent of the pathway involved in dendriticlength and complexity.

zDHHC15 promotes the trafficking of PSD-95 into dendritesAs zDHHC15 is known to palmitoylate PSD-95, and becauseexcitatory synapse density is reduced following zDHHC15knockdown, we tested whether PSD-95 palmitoylation is decreased

in zDHHC15 knockdown cells. Hippocampal neurons werenucleofected with zDHHC15 shRNA resulting in a 58±3.8%efficiency of transfection (investigated by counting the eGFP-positive transfected cells in each culture before lysis). By using theacyl-RAC assay, we demonstrated a 62.5±7.54% reduction inPSD-95 palmitoylation in primary neuron cultures transfected withzDHHC15 shRNA compared to control shRNA, with no change intotal PSD-95 levels (Fig. 4I,J).

Previous work has demonstrated that the N-terminalpalmitoylation motif of PSD-95 is necessary for targeting ofPSD-95 into dendrites (El-Husseini Ael et al., 2001), as well as itsassociation with vesiculotubular structures that traffic PSD-95 tosynapses (El-Husseini et al., 2000). To determinewhether zDHHC15is specifically involved in the targeting and trafficking of PSD-95,hippocampal neurons were transfected with PSD-95-RFP plus theconstructs indicated in Fig. 4K. All PSD-95 puncta within an entiredendrite were photobleached, and fluorescent recovery within thephotobleached dendrite was measured 9 h after photobleaching(Fig. 4K,L).While neurons transfected with control shRNA exhibited73±7% recovery of PSD-95-RFP fluorescence, neurons transfectedwith zDHHC15 shRNA exhibited only 35±6% recovery. Thisknockdown phenotype was rescued by co-transfecting neuronswith zDHHC15R (80±8% recovery) (Fig. 4K,L). In support of thefact that zDHHC15 regulates trafficking of PSD-95 into dendrites,PSD-95-RFP levels were significantly higher in the soma of neuronsexpressing shRNA targeting zDHHC15 (referred to in the figure asshRNA) (Fig. 4K).

Our data suggest that the decrease in excitatory synapse densitywithin zDHHC15 knockdown neurons is due to decreased PSD-95trafficking into dendrites. While the synapse density phenotype inknockdown cells could be rescued by zDHHC15, zDHHC3 andzDHHC2, all of which mediate PSD-95 palmitoylation (Fukataet al., 2004), the rescue with either zDHHC3 or zDHHC15 wassignificantly more robust than the rescue with zDHHC2. AszDHHC3 and zDHHC15 are both localized to the somatic Golgi(Greaves et al., 2011; Noritake et al., 2009), it is likely that these twoenzymes regulate excitatory synapse density by palmitoylatingPSD-95 and promoting the trafficking of PSD-95 into dendrites. Incontrast, zDHHC2 is localized to dendritic compartments (Greaveset al., 2011; Noritake et al., 2009), and has been shown to mediatePSD-95 palmitoylation and to promote its clustering at synapses(Noritake et al., 2009).

Together, our data demonstrate a role for zDHHC15 in theregulation of dendritic outgrowth, and the formation and maturationof glutamatergic synapses. This is of particular interest, given theassociation between zDHHC15 and X-linked intellectual disability(Mansouri et al., 2005; Piton et al., 2013), and the fact thatdisruptions in dendrite outgrowth and synapse function are one ofthe most-consistent hallmarks of intellectual disability, observed inboth postmortem brain sections and mouse models of intellectualdisability (Casanova et al., 2012; Huttenlocher, 1974; Jiang et al.,1998; Marin-Padilla, 1972; Purpura, 1974; Swann et al., 2000).

MATERIALS AND METHODSContact for reagent and resource sharingFurther information and requests for reagents may be directed to and will befulfilled by the corresponding author S.X.B.

AntibodiesPrimary antibodies used were: anti-zDHHC15 (Abcam, Cambridge, MA,#ab121203, 1:250, western blot), anti-zDHHC15 (Thermo Scientific,Waltham, MA, #PA39327, immunofluorescence, 1:100), anti-Flag(Sigma Aldrich, St Louis, MO, #F3165, 1:1000), anti-Myc (Origene,

5

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 6: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

Rockville, MD, #TA150121, 1:1000), anti-β-actin (Novus Biologicals,Centennial, CO, #NB600-503, 1:1000), anti-PSD-95 (Abcam,Cambridge, MA, #ab2723, 1:500), anti-gephyrin (Synaptic Systems,Göttingen, Germany #147011, 1:500), anti-GAD65 (SynapticSystems, Göttingen, Germany #198104, 1:500), anti-GFAP (SynapticSystems, Göttingen, Germany #173004, 1:400), anti-MAP2 (Millipore,Burlington, MA, MB3418, 1:2000), anti-giantin (Synaptic Systems,Göttingen, Germany, #263004, 1:500), anti-GM130 (BD Biosciences,

Franklin Lakes, NJ, #610822, 1:200). Secondary antibodies from LifeTechnologies (Carlsbad, CA,) used: (1:500), anti-mouse AF568 IgG2a(#A21134), anti-mouse AF647 IgG1 (#21240), anti-rabbit AF488(#A11008), anti-rabbit AF568 (#A11011), anti-guinea pig AF633(#A21105). HRP conjugated secondary antibodies were obtained fromBio-Rad (Hercules, CA): goat anti-mouse #170-6516 and goat anti-rabbit(#170-6515, 1:300). DAPI was used for nuclear staining (LifeTechnologies, Carlsbad, CA, #D1306, 1:1000).

Fig. 4. See next page for legend.

6

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 7: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

Plasmids and primersThe rat Myc-DDK tagged zDHHC15 ORF construct was obtainedfrom Origene (Rockville, MD, #RR212342). The shRNA target sequencethat gave maxium knockdown efficiency of zDHHC15 was 5′-GGTTCAATCTTGGCTTCATCA-3′ and was cloned into the pLL3.7vector (a gift from Luk Parijs; Addgene plasmid #11795) using thefollowing sense and anti-sense sequences based on previous cloningexperiments in pLL3.7 using XhoI and HpaI sites (Rubinson et al., 2003):Sense: 5′-TGGTTCAATCTTGGCTTCATCATTCAAGAGATGATGAA-GCCAAGATTGAACCTTTTTTC-3′ and antisense: 5′-TCGAGAAAAA-AGGTTCAATCTTGGCTTCATCATCTCTTGAATGATGAAGCCAAG-ATTGAACCA-3′. This plasmid is referred to as shRNA in all figures. Togenerate a scrambled shRNA construct to control for potential off-targeteffects of shRNA (control shRNA), we used the following sense and anti-sense sequence: Sense: 5′-TGAAGTTCGTACTTATCTCCGTTTCAAG-AGAACGGAGATAAGTACGAACTTCTTTTTTC-3′ and antisense: 5′-TCGAGAAAAAAGAAGTTCGT-ACTTATCTCCGTTCTCTTGAAAC-GGAGATAAGTACGAACTTCA-3′. To generate an shRNA-resistant formof zDHHC15 (zDHHC15R), site-directed mutagenesis (AgilentTechnologies, Santa Clara, CA: QuikChange II XL Site-DirectedMutagenesis Kit, 200521) of the zDHHC15 ORF sequenceGGTTCAATCTTGGCTTCATCA into GGTTTAATCTAGGATTCATAA

was carried out by using the following forward and reverse primers: Forward5′-CAGAGAAAAATGGGTTTAATCTAGGATTCATAAAGAATATTC-AG-3′ and Reverse 5′-CTGAATATTCTTTATGAATCCTAGATTAAAC-CCATTTTTCTCTG-3′ (underlined residues indicate amino acidcodons where nucleotide bases were mutated). zDHHS15R was made byusing the zDHHC15R template with the following forward and reverseprimers: Forward 5′-AAATGGACCATCATTCCCCATGGGTTA-AT-3′and Reverse 5′-ATTAACCCATGGG-GAATGATGGTCCATTT-3′.

Experimental model and subject detailsAnimalsAll experimental procedures and housing conditions were approved by theUBC Animal Care Committee and were in accordance with the CanadianCouncil on Animal Care (CCAC) guidelines.

Primary culture from Sprague-Dawley ratsHippocampi from embryonic day 18 (E18) Sprague-Dawley rats(Rattus norvegicus) of either sex were prepared and plated on 18 mmMarienfeld (Lauda-Königshofen, Germany) coverslips for 12-well plates ordirectly onto the plastic of 6-well plates at a density of 130 cells/mm2.Coverslips had previously been treated with concentrated nitric acid(Sigma Aldrich, St Louis, MO) overnight, rinsed several times with milliQwater, then sterilized with high heat (>250°C overnight). Coverslips werethen placed in the 12-well plate, UV sterilized for 30 min, and coated with0.5 mg/mL poly-L-lysine hydrobromide mixed with borate buffer. Theplates were covered with aluminium foil and left in a biosafety cabinetovernight. The following day, coverslips were rinsed 3× with autoclavedmilliQ water and plating medium (84.75 ml MEM with Earle’s BSS, 10 mlFBS, 2.25 ml of 20% glucose, 1 ml sodium pyruvate, 1 ml 100×GlutaMax,1 ml 100×Pen/Strep) was added (1 ml per 12 well, 2 ml per 6 well). Plateswere then placed in the 5% CO2 incubator at 37°C overnight.

For dissection, timed-pregnant Sprague-Dawley rats (Rattus norvegicus,JacksonLaboratories, Sacramento, CA)were euthanized usingCO2 and cervicaldislocation. Pups were harvested into 150-mm Petri dishes and decapitated, andthe brains were then dissected out and placed in pre-chilled HBSS on ice.Hippocampi were dissected and placed in a 15 ml conical tube with 10 ml offresh pre-warmed (37°C) HBSS. Once hippocampi settled, supernatant wasremoved and replaced with 10 ml of fresh pre-warmed HBSS for a total of threerinses. After the final rinse, 4.5 ml of fresh, pre-warmed HBSSwas added to thehippocampi with 0.5 ml of 2.5% trypsin and incubated in a 37°C water bath for20 min with gentle agitation every 5 min. In the final 3 min, 1% DNase wasadded and gently agitated to ensure mixture. Hippocampi were then rinsed 3×with fresh, prewarmedHBSS and, after the final wash,∼1 ml ofHBSSwas usedto transfer hippocampi to a 60-mm dish for trituration. The total volumefollowing trituration was brought up to 5 ml and cell density was determinedusing a hemocytometer to plate at the desired density. At 3-4 h after plating,plating medium was aspirated and replaced with pre-warmed (37°C)maintenance media (97 ml Neurobasal medium, 2 ml 50× B-27 supplement,1 ml 100×GlutaMax, 1 ml 100× Pen/Strep).Maintenancemediumwas renewedwith fresh maintenance medium 2–3 days following plating.

Method detailsTransfection (primary hippocampal cultures/HEK293T cells)Primary hippocampal cultures were transfected at 9–11 DIV by usingLipofectamine 2000 (Invitrogen, Carlsbad, CA) according to themanufacturer’s recommendations. Briefly, for 12-well-plates, two aliquotsof 25 μl Opti-Mem (Gibco, Thermo Fisher Scientific, Waltham, MA) wereprepared. To one aliquot, 1–3 μg of total plasmid DNA was added. To theother aliquot, 1 μl of Lipofectamine 2000 was added and allowed to mix for5 min. After this period, aliquots were combined and incubated for 20 min,then added drop by drop to the 12-well plates. Cells were then live imaged(10–13 DIV) or fixed (13 DIV) for subsequent experiments. HEK293T cellswere transfected with Lipofectamine 2000 (Invitrogen) at 70% confluency ata ratio of 3:1 (shRNA or scrambled shRNA to target plasmid) and incubatedfor 48–72 h before harvesting for biochemical analysis. For HEK293T cells,150 µl Opti-Mem were used with 6 µl Lipofectamine 2000.

Nucleofections for 6-well plate biochemical assays were performedimmediately before plating at 0 DIV by using an Amaxa Nucleofection Kit

Fig. 4. zDHHC15 knockdown decreases excitatory synapse density inhippocampal neurons and disrupts PSD-95 trafficking into dendrites.(A) Representative confocal images of rat hippocampal neurons at 13 DIVexpressing eGFP plus the indicated constructs. Control shRNA is a scrambledform of the shRNA targeting zDHHC15 (zDHHC15 shRNA, shortened toshRNA) to ensure there are no off-target effects of the shRNA. zDHHC15R is aform of zDHHC15 that has had mutations introduced to modify the sequencerecognized by the targeting shRNA to make it shRNA resistant. zDHHS15R isa construct wherein the catalytic cysteine has been modified to serine toreduce palmitoylation activity and is also resistant to the targeting shRNA.Immunostaining was for the excitatory post-synaptic marker, PSD-95 and theinhibitory post-synaptic marker gephyrin (masked as per Fig. S2E). Scale bar:10 µm. (B) The density of PSD-95-positive puncta is significantly decreased inzDHHC15 knockdown neurons, while the density of gephyrin-positive punctaremains unchanged. (C) This alters the ratio of excitatory:inhibitory synapsedensity being formed onto zDHHC15 mutant neurons. n=51 (control), 43(shRNA), 33 (shRNA+zDHHC15), 34 (shRNA+zDHHS15), 33 (zDHHC15)neurons, three cultures. (D) The zDHHC15 shRNA-mediated reduction inexcitatory synapse density (density of colocalized PSD-95 and excitatory pre-synaptic marker VGlut1 puncta) is rescued by coexpression of zDHHC15,zDHHC2 or zDHHC3. (E) The area of PSD-95 puncta is unchanged in cellsexpressing zDHHC15 shRNA but significantly increased compared to controlsin cells expressing zDHHC15 shRNA plus zDHHC2. (F,G) zDHHC15,zDHHC2 and zDHHC3 rescue zDHCH15 shRNA-mediated changes in theproportion of filopodia (F) andmature spines (G). (H) By contrast, zDHHC2 andzDHHC3 are unable to rescue zDHHC15-mediated decreases in dendriticlength. n=25 neurons, three cultures. (I) Acyl-RAC assay from cultured neuronsat 7 DIV that were nucleofected with control or zDHHC15 shRNA. Left panel(input levels): zDHHC15 was reduced by 66.25±6% in neurons that had beentransfected with zDHHC15 shRNA, reflecting the efficiency of transfection. Nochanges in PSD-95 levels were observed. Right panel (Acyl-RAC fractions):Results from the same blot; PSD-95 palmitoylation is decreased in culturestransfected with zDHHC15 shRNA (cleaved bound fraction, cBF), low levels ofnon-palmitoylated PSD-95 (cUF, cleaved unbound fraction), and minimal non-specific binding to the resin (pBF, preserved bound fraction). The pUF(preserved unbound fraction) represents samples that have not been cleaved.(J) PSD-95 palmitoylation is decreased in zDHHC15 knockdown neurons. n=3cultures. ***P<0.001, Student’s t-test, mean±s.e.m. (K) Hippocampal neuronsat 10 DIV were transfected with the indicated constructs plus PSD-95-RFP.Puncta of PSD-95-RFP within an entire primary dendrite branch (white outline)were photobleached at 13 DIV and fluorescence recovery was analysed 9 hafter bleaching. Fluorescence recovery was decreased in cells expressingzDHHC15 shRNA (PSD-95 puncta pseudo-colored for fluorescenceintensity). Magnified area of interest shown below. Scale bar: 10 µm.(L) Quantification of fluorescence recovery. n=10 neurons, three cultures.***/###P<0.001, **/##P<0.01, *P<0.05, one-way ANOVA and Tukey’s post-hoctest, mean±s.e.m.

7

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 8: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

(Lonza, Basel, Switzerland, VPG-1003) according to the manufacturer’soptimized protocol (Number 101, program G-13), and were used forexperiments at either 4 DIV (Fig. 2A) or 7 DIV (Fig. 4I). Briefly,hippocampi were dissociated as above and ∼500,000 neurons isolated fornucleofection. Neurons were mixed with 100 μl Ingenio electroporationsolution (Mirus Bio, Madison, WI) and cDNA constructs (∼3 μg in total)were added to the solution. The solution was transferred to an AmaxaNucleofection cuvette and inserted into the Amaxa Nucleofector withprogram G-013. Following electroporation, the solution was plated into a 6-well dish for future biochemical experiments.

ImmunocytochemistryImmunocytochemistry experiments were performed as previously reported(Sun and Bamji, 2011). Briefly, cells were fixed for 10 min in a pre-warmed(37°C) 4% paraformaldehyde–sucrose solution. Cells were then washed inphosphate buffered saline (PBS) and treated with 0.1% Triton X-100 in PBSfor 10 min at room temperature, washed in PBS, and blocked for 1 h at roomtemperature with 10% goat serum in PBS. Cells were then incubated in 1%goat serum in PBS with primary antibodies overnight at 4°C. Subsequently,cells were washed 3× in PBS for 10 min each, incubated in secondaryantibodies in 1% goat serum in PBS for 1 h at RT, washed again 3× in PBSand mounted on microscope slides by using Prolong Gold (MolecularProbes, Thermo Fisher Scientific, Waltham, MA). For DAPI staining, a1:100 dilution from a 5 mg/ml stock was applied after all other stainingprocedures for 5 min. Cells were then washed 3× in PBS and then mountedas above.

BiotinylationFor surface biotinylation assays, neurons in 6-well plates were washed withice-cold PBS pH 8 supplemented with 0.1 mM CaCl2 and 1 mM MgCl2(PBS-CM) and then incubated for 30 min with 0.5 mg/ml NHS-SS-Biotin(Thermo Fisher Scientific, Waltham, MA) in ice-cold PBS-CM at 4°C withgentle rocking. After incubation, cells were washed once with PBS-CM; theunbound biotin was then quenched during two 8 min incubations withquenching buffer (20 mM glycine in PBS-CM). Lysis was performed bymechanical scraping in lysis buffer (1% IGEPAL-CA630 and 1 mM PMSFwith Roche complete protease inhibitor tablet) and followed by centrifugationat 500 g for 5 min at 4°C. Samples were then vortexed, run 3–4 times througha 26½ gauge syringe, and nutated at 4°C (VWRNutatingMixer, 20° fixed tiltangle with a mixing speed of 24 rpm) for 30 min. After nutation, sampleswere centrifuged at 16,100 g for 30 min at 4°C to clear the lysate. Proteincontent in the cell lysate was then quantified using a BCAAssay Kit (ThermoFisher Scientific, Waltham, MA) as per the manufacturer’s instructions.Whole-cell lysate (10 µg) was then combined with SDS-sample buffer(50 mM Tris-HCl, 2% SDS, 10% glycerol, 14.5 mM EDTA and 0.02%Bromophenol Blue with 1% β-mercaptoethanol), boiled for 5 min at 95°Cand stored at −20°C as the input sample. Of the remaining protein sample,100–200 µg was added to 50 µl of a 50% slurry of NeutrAvidin-conjugatedagarose beads (Thermo Fisher Scientific, Waltham, MA) that had been pre-washed three times in lysis buffer. Each sample was then brought to a totalvolume of 500 µl with lysis buffer and nutated (VWR Nutating Mixer, 20°fixed tilt angle with a mixing speed of 24 rpm) overnight at 4°C. Thefollowing day, beads were pelleted and washed seven times by centrifugationat 500 g for 3 min, each time discarding the supernatant. Beads were elutedusing 40 µl of SDS-sample buffer supplemented with 100 mM DTT andsamples were boiled at 90°C for 5 min followed by western blotting withwhole-cell lysates.

Acyl-RAC (palmitoylation) assayFor Acyl-RAC assays, we followed the manufacturer’s protocol from theCAPTUREome S-palmitoylated protein kit (Badrilla, Leeds, UK), withminor changes. We optimized the protocol for our experiments by addingDNase to the lysed-cell solution. Additionally, we measured proteinconcentration (BCA Assay, Thermo Fisher Scientific, Waltham, MA) afterdissolving the precipitated protein and prior to separating the lysate intoexperimental (Thioester Cleavage Reagent) and negative control (AcylPreservation Reagent) samples to ensure accurate loading of equal proteinconcentrations.

Western blot analysisWestern blotting was performed as previously described (Sun and Bamji,2011). Primary hippocampal neurons from Sprague-Dawley rats, HEK293Tcells, or brain regions from mouse (i.e. cortex or cerebellum) were lysed atthe time points indicated in figures (Fig. 1A, mouse brain regions, E17, P10,P90; Fig. 1D, hippocampal neurons, 13 DIV; Fig. 2A, Fig. S4, hippocampalneurons, 4 DIV; Fig. 4I, hippocampal neurons, 7 DIV; Fig. S1A, HEK293Tcells, 95–100% confluency) in ice-cold RIPA buffer or lysis buffer (1%IGEPAL CA-630, 50 mM Tris-HCl-pH 7.5, 150 mM NaCl, 10% glycerol)supplemented with protease inhibitor cocktail solution at 4°C on a nutator(VWR Nutating Mixer) for 2 h at 20° fixed tilt and 24 rpm. Lysates werecleared by benchtop centrifugation (16,000 g) for 30 min at 4°C and thesupernatant was mixed with 5×SDS dye for SDS-PAGE. Proteins wereseparated on either a 10% resolving gel or a 4–20% gradient gel, transferredonto a PVDF membrane, blocked in 3% BSA (in TBST) and were thenimmunoblotted for were then immunoblotted for zDHHC15 (Fig. 1A,D,Fig. 2A, Fig. 4I; Fig. S1A), β-actin (Fig. 1A, Fig. 2A, Fig. 4I; Fig. S1A,Fig. S4), zDHHC5 (Fig. 1D), zDHHC8 (Fig. 1D), PSD-95 (Fig. 4I) or Myc(Fig. S1A, Fig. S4) (prepared in 0.3% BSA in TBST) and visualized usingenhanced chemiluminiscence reagent (MerckMillipore, #WBKLS0500) ona Li-Cor C-Digit Blot Scanner (LI-COR, Lincoln, NE). Blots werequantified using ImageJ software (NIH, Bethesda, Maryland).

ImagingFixed and live neuronswere imaged by using an invertedOlympus (RichmondHill, ONT, Canada) Fluoview 1000 (FV1000) confocal microscope. Imagingof synapses and colocalization studies utilized the 60×/1.42 Oil Plan-Apochromat objective while dendritic length measurements utilized the 20×/0.75 Oil Plan-Apochromat objective. MatTek glass-bottomed culture dishes(P35G-1.5-14-C, MatTek Corporation, Ashland, MA) were used to performlive imaging of dendrite growth of neurons in a chamber maintained at 37°C.eGFP-transfected cells with different experimental conditions were imaged inHEPES buffer (140 mM NaCl, 1.3 mM CaCl2, 1.3 mM MgCl2, 2.4 mMK2HPO4, 25 mM HEPES pH 7.4) every 24 h post transfection up to 72 h toensure cell viability. Levels and contrast of confocal images were moderatelyadjusted in Photoshop CS6 software (Adobe Systems, San Jose, CA) by usingscientifically acceptable procedures.

Fluorescence recovery after photobleachingFluorescence recovery after photobleaching (FRAP) experiments wereperformed as previously described (Brigidi et al., 2014, 2015) but updatedfor use on an Olympus (Richmond Hill, ONT, Canada) FV3000microscope. Briefly, cells transfected with eGFP and PSD-95 wereidentified and up to four dendritic branches of the cell that were clear ofother obstructions and relatively unbranched were photobleached from thesoma to the end of the dendrite. Cell positions were recorded using themotorized stage and ‘positions’ function of the FV3000, and re-imaged 9 hafter bleaching. Fluorescence recovery was quantified using processingsoftware within the Olympus FV3000 and then analyzed in Prism software(Graphpad, La Jolla, CA).

Image analysis and quantificationDendrite length and Sholl analysisNeurons at 10–11 DIV were transfected and fixed at DIV 13–14. To measuredendrite lengths, neurons were identified through eGFP fluorescence andimaged at a magnification of 20×. Images were processed using AdobePhotoshop to remove axons. Imageswere converted to 8-bit and then exportedinto the ImageJ tool-NeuronJ (Meijering et al., 2004) to trace the dendriticlength. Sholl analysis (dendritic complexity analysis) was performed on thesame 20× images that were first thresholded and then processed using the‘Sholl analysis’ plugin from ImageJ (Ferreira et al., 2014) with settings tocreate a step size of 2 µm and an ending radius entered as 'NaN' (not a number)to enable sampling of the entire image to ensure the entire dendritic tree wascaptured; see Fig. S3B. Data were imported into Microsoft Excel to create anaverage Sholl profile for each condition. For all images, the scale for ImageJwas set using the raw data from the Olympus FV1000/FV3000 dimensions todetermine µm per pixel.

8

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 9: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

Spine analysisDendritic spines were classified by using the default values and analysisfrom NeuronStudio (version 0.9.92) adapted from Rodriguez et al. (2006),which uses a classification scheme based on the head to neck diameterratio, length to head diameter ratio and the head diameter. For dataanalysis, stubby and mushroom-type spines were classified as maturespines, whereas filopodial and/or thin protrusions were considered asimmature spines.

Synapse density, puncta size analysis and colocalizationTransfected neurons were immunostained for PSD-95 (as a marker ofexcitatory synapses) and gephyrin (as a marker for inhibitory synapses).Synapses within eGFP-transfected neurons were identified by making aneuronal ‘mask’ created from the GFP-cell fill, imaged at 60×magnification, in Adobe Photoshop CS6 or CC2017 (Adobe Systems,San Jose, CA). The mask was devoid of cell soma and axon. We used amagic wand tool (non-contiguous, tolerance 30) to select a blackbackground and this cell-fill GFP mask was applied onto PSD-95-positiveand/or gephyrin-positive synapse marker channels to exclude synapses fromother untransfected neurons and only examine synapses that had formed onmutant cells. These masks from synapse channels were then manuallythresholded to make binary images and the total number of synaptic punctawas quantified using a macro in order to define synaptic puncta as having asize between 0.05 µm2 and 3 µm2. To obtain synapse density, the totalnumber of puncta was divided by the length of the masked dendrite (see Fig.S1B). For the puncta size measurements, masked images were subjectivelythresholded and PSD-95-positive puncta were measured for Feret’sdiameter, and then averaged for each image under each condition. Maskedimages were then thresholded, and the ‘Colocalization’ plugin (https://imagej.nih.gov/ij/plugins/colocalization.html) was used in a custom macrowhen applying the ‘Analyze Particles’ function of ImageJ (NIH, Bethesda,MA) to determine colocalized points from separate channels.

Statistical analysisFor all experiments, data represent the mean±s.e.m. Statistical significancewas measured using ANOVA with Tukey’s multiple comparison analysisin Graphpad Prism v6.01 (Graphpad, La Jolla, CA). For all imagingexperiments, the n value is the number of cells used per condition, for at leastthree independent cultures. Statistical significance is assumed when α<0.05,unless mentioned otherwise (Figs 3E,F and 4F,G, where the α-value is mademore-stringent to 0.001). Significance is indicated when */#P<0.05,**/##P<0.01 or ***/###P<0.001 using one-way ANOVA, Tukey’s multiplecomparison test with mean ±s.e.m., and determined by Prism software. Allfigures were generated by using Adobe Illustrator CS6 or CC2018 softwarein conjunction with Adobe Photoshop CS6 or CC2017 (Adobe Systems,San Jose, CA). Statistical significances (*) indicate significant differencescompared with the control; significance within different rescue constructs isindicated by #.

AcknowledgementsThe authors thank all members of the Bamji lab for critical review of the manuscript.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsConceptualization: S.X.B.; Formal analysis: B.S.S., J.J.S.; Investigation: B.S.S.,J.J.S.; Data curation: B.S.S., J.J.S.; Writing - original draft: B.S.S., J.J.S., S.X.B.;Writing - review & editing: B.S.S., J.J.S., S.X.B.; Visualization: B.S.S., J.J.S.;Supervision: S.X.B.; Project administration: S.X.B.; Funding acquisition: S.X.B.

FundingThe work was supported by the Canadian Institutes of Health Research(MOP-142721 and PJT-148938) to S.X.B. Deposited in PMC for immediate release.

Supplementary informationSupplementary information available online athttp://jcs.biologists.org/lookup/doi/10.1242/jcs.230052.supplemental

ReferencesBrigidi, G. S., Santyr, B., Shimell, J., Jovellar, B. andBamji, S. X. (2015). Activity-

regulated trafficking of the palmitoyl-acyl transferase DHHC5. Nat. Commun. 6,8200. doi:10.1038/ncomms9200

Brigidi, G. S., Sun, Y., Beccano-Kelly, D., Pitman, K., Mobasser, M., Borgland,S. L., Milnerwood, A. J., and Bamji, S. X. (2014). Palmitoylation of δ-catenin byDHHC5 mediates activity-induced synapse plasticity. Nat. Neurosci. 17, 522-532.doi:10.1038/nn.3657

Casanova, J. R., Nishimura, M., Owens, J. W. and Swann, J. W. (2012). Impact ofseizures on developing dendrites: implications for intellectual developmentaldisabilities. Epilepsia 53 Suppl. 1, 116-124. doi:10.1111/j.1528-1167.2012.03482.x

Charollais, J. andVanDerGoot, F. G. (2009). Palmitoylation ofmembrane proteins(Review). Mol. Membr. Biol. 26, 55-66. doi:10.1080/09687680802620369

El-Husseini, A. E., Craven, S. E., Chetkovich, D. M., Firestein, B. L., Schnell, E.,Aoki, C. and Bredt, D. S. (2000). Dual palmitoylation of PSD-95 mediates itsvesiculotubular sorting, postsynaptic targeting, and ion channel clustering. J. CellBiol. 148, 159-172. doi:10.1083/jcb.148.1.159

El-Husseini Ael, D., Craven, S. E., Brock, S. C. and Bredt, D. S. (2001). Polarizedtargeting of peripheral membrane proteins in neurons. J. Biol. Chem. 276,44984-44992. doi:10.1074/jbc.M103049200

Fang, C., Deng, L., Keller, C. A., Fukata, M., Fukata, Y., Chen, G. and Luscher, B.(2006). GODZ-mediated palmitoylation of GABA(A) receptors is required fornormal assembly and function of GABAergic inhibitory synapses. J. Neurosci. 26,12758-12768. doi:10.1523/JNEUROSCI.4214-06.2006

Ferreira, T., Ou, Y., Li, S., Giniger, E. and van Meyel, D. J. (2014). Dendritearchitecture organized by transcriptional control of the F-actin nucleator Spire.Development 141, 650-660. doi:10.1242/dev.099655

Fukata, Y. and Fukata, M. (2010). Protein palmitoylation in neuronal developmentand synaptic plasticity. Nat. Rev. Neurosci. 11, 161-175. doi:10.1038/nrn2788

Fukata, M., Fukata, Y., Adesnik, H., Nicoll, R. A. and Bredt, D. S. (2004).Identification of PSD-95 palmitoylating enzymes. Neuron 44, 987-996. doi:10.1016/j.neuron.2004.12.005

Greaves, J., Salaun, C., Fukata, Y., Fukata, M. and Chamberlain, L. H. (2008).Palmitoylation and membrane interactions of the neuroprotective chaperonecysteine-string protein. J. Biol. Chem. 283, 25014-25026. doi:10.1074/jbc.M802140200

Greaves, J., Gorleku, O. A., Salaun, C. and Chamberlain, L. H. (2010).Palmitoylation of the SNAP25 protein family: specificity and regulation byDHHC palmitoyl transferases. J. Biol. Chem. 285, 24629-24638. doi:10.1074/jbc.M110.119289

Greaves, J., Carmichael, J. A. and Chamberlain, L. H. (2011). The palmitoyltransferase DHHC2 targets a dynamicmembrane cycling pathway: regulation by aC-terminal domain. Mol. Biol. Cell 22, 1887-1895. doi:10.1091/mbc.e10-11-0924

Huttenlocher, P. R. (1974). Dendritic development in neocortex of children withmental defect and infantile spasms. Neurology 24, 203-210. doi:10.1212/WNL.24.3.203

Jiang, M., Lee, C. L., Smith, K. L. and Swann, J. W. (1998). Spine loss and otherpersistent alterations of hippocampal pyramidal cell dendrites in a model of early-onset epilepsy. J. Neurosci. 18, 8356-8368. doi:10.1523/JNEUROSCI.18-20-08356.1998

Ko, P. J. andDixon, S. J. (2018). Protein palmitoylation and cancer.EMBORep. 19,e466666. doi:10.15252/embr.201846666

Levy, A. D., Devignot, V., Fukata, Y., Fukata, M., Sobel, A. and Chauvin, S.(2011). Subcellular Golgi localization of stathmin family proteins is promoted by aspecific set of DHHC palmitoyl transferases.Mol. Biol. Cell 22, 1930-1942. doi:10.1091/mbc.e10-10-0824

Linhares, N. D., Valadares, E. R., da Costa, S. S., Arantes, R. R., de Oliveira,L. R., Rosenberg, C., Vianna-Morgante, A. M. and Svartman, M. (2016).Inherited Xq13.2-q21.31 duplication in a boy with recurrent seizures and pubertalgynecomastia: Clinical, chromosomal and aCGH characterization. Meta Gene 9,185-190. doi:10.1016/j.mgene.2016.07.004

Mansouri, M. R., Marklund, L., Gustavsson, P., Davey, E., Carlsson, B.,Larsson, C., White, I., Gustavson, K. H. and Dahl, N. (2005). Loss of ZDHHC15expression in a woman with a balanced translocation t(X;15)(q13.3;cen) andsevere mental retardation. Eur. J. Hum. Genet. 13, 970-977. doi:10.1038/sj.ejhg.5201445

Marin-Padilla, M. (1972). Structural abnormalities of the cerebral cortex in humanchromosomal aberrations: a Golgi study. Brain Res. 44, 625-629. doi:10.1016/0006-8993(72)90324-1

Martinez, F., Rosello, M., Mayo, S., Monfort, S., Oltra, S. and Orellana, C. (2014).Duplication at Xq13.3-q21.1 with syndromic intellectual disability, a probable rolefor the ATRX gene. Am. J. Med. Genet. 164A, 918-923. doi:10.1002/ajmg.a.36371

Meijering, E., Jacob, M., Sarria, J. C., Steiner, P., Hirling, H. and Unser, M.(2004). Design and validation of a tool for neurite tracing and analysis influorescence microscopy images. Cytometry A 58, 167-176. doi:10.1002/cyto.a.20022

Mill, P., Lee, A. W., Fukata, Y., Tsutsumi, R., Fukata, M., Keighren, M., Porter,R. M., McKie, L., Smyth, I. and Jackson, I. J. (2009). Palmitoylation regulates

9

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience

Page 10: Regulation of dendrite morphology and excitatory synapse ... · University of British Columbia, 2350 Health Sciences Mall, Vancouver, B.C., V6T-1Z3, Canada. *These authors contributed

epidermal homeostasis and hair follicle differentiation. PLoS Genet. 5, e1000748.doi:10.1371/journal.pgen.1000748

Noritake, J., Fukata, Y., Iwanaga, T., Hosomi, N., Tsutsumi, R., Matsuda, N.,Tani, H., Iwanari, H., Mochizuki, Y., Kodama, T. et al. (2009). Mobile DHHCpalmitoylating enzyme mediates activity-sensitive synaptic targeting of PSD-95.J. Cell Biol. 186, 147-160. doi:10.1083/jcb.200903101

Ohno, Y., Kihara, A., Sano, T. and Igarashi, Y. (2006). Intracellular localization andtissue-specific distribution of human and yeast DHHC cysteine-rich domain-containing proteins. Biochim. Biophys. Acta 1761, 474-483. doi:10.1016/j.bbalip.2006.03.010

Piton, A., Redin, C. and Mandel, J.-L. (2013). XLID-causing mutations andassociated genes challenged in light of data from large-scale human exomesequencing. Am. J. Hum. Genet. 93, 368-383. doi:10.1016/j.ajhg.2013.06.013

Purpura, D. P. (1974). Dendritic spine “dysgenesis” andmental retardation.Science186, 1126-1128. doi:10.1126/science.186.4169.1126

Rocks, O., Gerauer, M., Vartak, N., Koch, S., Huang, Z.-P., Pechlivanis, M.,Kuhlmann, J., Brunsveld, L., Chandra, A., Ellinger, B. et al. (2010). Thepalmitoylationmachinery is a spatially organizing system for peripheral membraneproteins. Cell 141, 458-471. doi:10.1016/j.cell.2010.04.007

Rodriguez, A., Ehlenberger, D. B., Hof, P. R. and Wearne, S. L. (2006). Rayburstsampling, an algorithm for automated three-dimensional shape analysis fromlaser scanning microscopy images.Nat. Protoc. 1, 2152-2161. doi:10.1038/nprot.2006.313

Rubinson, D. A., Dillon, C. P., Kwiatkowski, A. V., Sievers, C., Yang, L., Kopinja,J., Rooney, D. L., Zhang, M., Ihrig, M. M., McManus, M. T. et al. (2003). Alentivirus-based system to functionally silence genes in primary mammalian cells,stem cells and transgenic mice by RNA interference. Nat. Genet. 33, 401-406.doi:10.1038/ng1117

Sanders, S. S., Martin, D. D., Butland, S. L., Lavallee-Adam, M., Calzolari, D.,Kay, C., Yates, J. R., III and Hayden, M. R. (2015). Curation of the mammalianpalmitoylome indicates a pivotal role for palmitoylation in diseases and disorders

of the nervous system and cancers. PLoS Comput. Biol. 11, e1004405. doi:10.1371/journal.pcbi.1004405

Sharma, C., Yang, X. H. and Hemler, M. E. (2008). DHHC2 affects palmitoylation,stability, and functions of tetraspanins CD9 and CD151. Mol. Biol. Cell 19,3415-3425. doi:10.1091/mbc.e07-11-1164

Sun, Y. and Bamji, S. X. (2011). beta-Pix modulates actin-mediated recruitment ofsynaptic vesicles to synapses. J. Neurosci. 31, 17123-17133. doi:10.1523/JNEUROSCI.2359-11.2011

Swann, J. W., Al-Noori, S., Jiang, M. and Lee, C. L. (2000). Spine loss and otherdendritic abnormalities in epilepsy.Hippocampus 10, 617-625. doi:10.1002/1098-1063(2000)10:5<617::AID-HIPO13>3.0.CO;2-R

Tsutsumi, R., Fukata, Y., Noritake, J., Iwanaga, T., Perez, F. and Fukata, M.(2009). Identification of G protein alpha subunit-palmitoylating enzyme.Mol. Cell.Biol. 29, 435-447. doi:10.1128/MCB.01144-08

Vetrivel, K. S., Meckler, X., Chen, Y., Nguyen, P. D., Seidah, N. G., Vassar, R.,Wong, P. C., Fukata, M., Kounnas, M. Z. and Thinakaran, G. (2009). Alzheimerdisease Abeta production in the absence of S-palmitoylation-dependenttargeting of BACE1 to lipid rafts. J. Biol. Chem. 284, 3793-3803. doi:10.1074/jbc.M808920200

Wang, F., Chen, X., Shi, W., Yao, L., Gao, M., Yang, Y. and Hao, A. (2015).Zdhhc15b regulates differentiation of diencephalic dopaminergic neurons inzebrafish. J. Cell. Biochem. 116, 2980-2991. doi:10.1002/jcb.25256

Yokoi, N., Fukata, Y., Sekiya, A., Murakami, T., Kobayashi, K. and Fukata, M.(2016). Identification of PSD-95 depalmitoylating enzymes. J. Neurosci. 36,6431-6444. doi:10.1523/JNEUROSCI.0419-16.2016

Zhang, Y., Chen, K., Sloan, S. A., Bennett, M. L., Scholze, A. R., O’Keeffe, S.,Phatnani, H. P., Guarnieri, P., Caneda, C., Ruderisch, N. et al. (2014). An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascularcells of the cerebral cortex. J. Neurosci. 34, 11929-11947. doi:10.1523/JNEUROSCI.1860-14.2014

10

SHORT REPORT Journal of Cell Science (2019) 132, jcs230052. doi:10.1242/jcs.230052

Journal

ofCe

llScience


Recommended