6
AMPA receptor and GEF-H1/Lfc complex regulates dendritic spine development through RhoA signaling cascade Myoung-Goo Kang a,1 , Yurong Guo b , and Richard L. Huganir a,2 a Department of Neuroscience, The Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205; and b Department of Medicine, The Johns Hopkins University, 5200 Eastern Avenue, Baltimore, MD 21234 Contributed by Richard L. Huganir, December 19, 2008 (sent for review November 21, 2008) AMPA receptors (AMPA-R) are major mediators of synaptic trans- mission and plasticity in the developing and adult central nervous system. Activity-dependent structural plasticity mediated by dy- namic changes in the morphology of spines and dendrites is also essential for the formation and tuning of neuronal circuits. RhoA and Rac1 are known to play important roles in the regulation of spine and dendrite development in response to neuronal activity. These Rho GTPases are activated by guanine nucleotide exchange factors (GEFs). In this study, we identified GEF-H1/Lfc as a compo- nent of the AMPA-R complex in the brain. GEF-H1 is enriched in the postsynaptic density and is colocalized with GluR1 at spines. GEF-H1 activity negatively regulates spine density and length through a RhoA signaling cascade. In addition, AMPA-R-dependent changes in spine development are eliminated by down-regulation of GEF-H1. Altogether, these results strongly suggest that GEF-H1 is an important mediator of AMPA-R activity-dependent structural plasticity in neurons. glutamate receptor GTPase learning and memory structural plasticity synaptic plasticity D endritic spines are small actin-rich protrusions from neuronal dendrites with a globular head and thin neck. As a basic functional unit of the excitatory synapse, dendritic spines are critical for most excitatory synaptic transmission in the brain. Because dynamic changes in the shape, size, and number of spines are major forms of structural synaptic plasticity, the molecular mechanism underlying the regulation of spine development, maintenance, and dynamics has been an active research area in neuroscience. Spines are rich in F-actin and actin dynamics modulate the morphological plasticity of spines. In various cell types including neurons, actin dynamics is regulated by Rho GTPases in response to external cues (1). The Rho family of small GTPase consists of a large number of proteins including Rho, Rac, and Cdc42. These proteins are binary switches that cycle between GDP-bound inactive and a GTP-bound active state. In response to various extracellular signals, this switch is turned on or off by regulatory proteins. In neurons, Rho GTPases have been implicated in the cytoskeletal dynamics for structural plasticity of excitatory synapses. Particularly, RhoA and Rac1 have been known as key players for the regulation of spine and dendrite development and dynamics (2). The regulatory proteins of the Rho GTPases include guanine nucleotide exchange factors (GEFs), GTPase activating proteins, and guanine nucleotide dissociation inhibitors. GEFs catalyze the exchange of GDP for GTP to generate the active state of Rho GTPases. In contrast, GTPase activating proteins and guanine nucleotide dissociation inhibitors inactivate Rho GTPases. The expression of GEF proteins is tissue-specific, providing a molecular mechanism for tissue-specific modulation of Rho GTPases (3). Tiam1 is known as a neuronal GEF involved in NMDA receptor activity-dependent structural plasticity (4). However, neuronal GEFs involved in AMPA receptor (AMPA-R) activity-dependent structural plasticity have not been reported. Modulation of the trafficking and activity of AMPA-R by NMDA receptor activity is a major mechanism for synaptic plas- ticity (5). Recent studies have identified several AMPA-R inter- acting proteins that can modulate the activity and trafficking of AMPA-Rs (5). In this study, we immunopurified the AMPA-R from rat brain and found that the RhoA GEF, GEF-H1, is a significant component of the AMPA-R complex in vivo. Truncated GEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involved in cell proliferation (6–8). The full-length GEF-H1 was found as a 120-kDa protein consisting of 985 aa (9). Interestingly, GEF-H1 binding to microtubule regulates its enzymatic activity, suggesting GEF-H1 could mediate cross-talk between microtubules and actin (9, 10). Our functional analysis of GEF-H1 demonstrated that GEF-H1 negatively regulates the density and length of spines through a RhoA signaling cascade. In addition, AMPA-R activity- dependent changes in spine development were eliminated by inhi- bition of GEF-H1. Together, this study strongly suggests that GEF-H1 is an AMPA-R associated protein that is important for activity-dependent structural plasticity. Results Identification of GEF-H1 as a Component of the AMPA-R Complex. Through proteomic screening of AMPA-R-binding proteins in rat brains, GEF-H1, a GEF, was identified as a component of the AMPA-R complex in the brain [Fig. 1 and supporting information (SI) Fig. S1]. AMPA-R complexes were purified from rat brains using wheat germ agglutinin (WGA)-chromatography and large- scale immunoprecipitation (IP). WGA-chromatography was adapted to enrich for glycosylated mature membrane proteins and to exclude immature AMPA-R complexes in the endoplasmic reticulum and Golgi apparatus. To distinguish specific binding from nonspecific binding of proteins to IgG, Protein A, and Sepharose during the large-scale IP, the eluant from WGA-chromatography was divided into 2 pools and 1 pool was used as a negative control. In the negative control group, the GluR1 antibody was blocked with the synthetic peptide used for the generation of the antibody (see Fig. 1 A, Pep ). The isolation of many of the copurifying proteins was inhibited by preabsorbing the GluR1 antibody with the anti- Author contributions: M.-G.K. and R.L.H. designed research; M.-G.K. and Y.G. performed research; M.-G.K. and Y.G. contributed new reagents/analytic tools; M.-G.K., Y.G., and R.L.H. analyzed data; and M.-G.K. and R.L.H. wrote the paper. Conflict of interest statement: Under a licensing agreement between Millipore and the Johns Hopkins University, R.L.H. is entitled to a share of royalty received by the university on sales of products described in this article. R.L.H. is a paid consultant to Millipore. The terms of this arrangement are being managed by the Johns Hopkins University in accor- dance with its conflict of interest policies. Freely available online through the PNAS open access option. 1 Present address: Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555. 2 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/cgi/content/full/ 0812861106/DCSupplemental. © 2009 by The National Academy of Sciences of the USA www.pnas.orgcgidoi10.1073pnas.0812861106 PNAS March 3, 2009 vol. 106 no. 9 3549 –3554 NEUROSCIENCE Downloaded by guest on May 25, 2020

AMPA receptor and GEF-H1/Lfc complex regulates dendritic ... · GEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involved in cell proliferation (6–8). The full-length GEF-H1

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: AMPA receptor and GEF-H1/Lfc complex regulates dendritic ... · GEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involved in cell proliferation (6–8). The full-length GEF-H1

AMPA receptor and GEF-H1/Lfc complex regulatesdendritic spine development through RhoAsignaling cascadeMyoung-Goo Kanga,1, Yurong Guob, and Richard L. Huganira,2

aDepartment of Neuroscience, The Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, 725 North Wolfe Street,Baltimore, MD 21205; and bDepartment of Medicine, The Johns Hopkins University, 5200 Eastern Avenue, Baltimore, MD 21234

Contributed by Richard L. Huganir, December 19, 2008 (sent for review November 21, 2008)

AMPA receptors (AMPA-R) are major mediators of synaptic trans-mission and plasticity in the developing and adult central nervoussystem. Activity-dependent structural plasticity mediated by dy-namic changes in the morphology of spines and dendrites is alsoessential for the formation and tuning of neuronal circuits. RhoAand Rac1 are known to play important roles in the regulation ofspine and dendrite development in response to neuronal activity.These Rho GTPases are activated by guanine nucleotide exchangefactors (GEFs). In this study, we identified GEF-H1/Lfc as a compo-nent of the AMPA-R complex in the brain. GEF-H1 is enriched in thepostsynaptic density and is colocalized with GluR1 at spines.GEF-H1 activity negatively regulates spine density and lengththrough a RhoA signaling cascade. In addition, AMPA-R-dependentchanges in spine development are eliminated by down-regulationof GEF-H1. Altogether, these results strongly suggest that GEF-H1is an important mediator of AMPA-R activity-dependent structuralplasticity in neurons.

glutamate receptor � GTPase � learning and memory �structural plasticity � synaptic plasticity

Dendritic spines are small actin-rich protrusions from neuronaldendrites with a globular head and thin neck. As a basic

functional unit of the excitatory synapse, dendritic spines are criticalfor most excitatory synaptic transmission in the brain. Becausedynamic changes in the shape, size, and number of spines are majorforms of structural synaptic plasticity, the molecular mechanismunderlying the regulation of spine development, maintenance, anddynamics has been an active research area in neuroscience. Spinesare rich in F-actin and actin dynamics modulate the morphologicalplasticity of spines. In various cell types including neurons, actindynamics is regulated by Rho GTPases in response to externalcues (1).

The Rho family of small GTPase consists of a large number ofproteins including Rho, Rac, and Cdc42. These proteins are binaryswitches that cycle between GDP-bound inactive and a GTP-boundactive state. In response to various extracellular signals, this switchis turned on or off by regulatory proteins. In neurons, Rho GTPaseshave been implicated in the cytoskeletal dynamics for structuralplasticity of excitatory synapses. Particularly, RhoA and Rac1 havebeen known as key players for the regulation of spine and dendritedevelopment and dynamics (2).

The regulatory proteins of the Rho GTPases include guaninenucleotide exchange factors (GEFs), GTPase activating proteins,and guanine nucleotide dissociation inhibitors. GEFs catalyze theexchange of GDP for GTP to generate the active state of RhoGTPases. In contrast, GTPase activating proteins and guaninenucleotide dissociation inhibitors inactivate Rho GTPases. Theexpression of GEF proteins is tissue-specific, providing a molecularmechanism for tissue-specific modulation of Rho GTPases (3).Tiam1 is known as a neuronal GEF involved in NMDA receptoractivity-dependent structural plasticity (4). However, neuronalGEFs involved in AMPA receptor (AMPA-R) activity-dependentstructural plasticity have not been reported.

Modulation of the trafficking and activity of AMPA-R byNMDA receptor activity is a major mechanism for synaptic plas-ticity (5). Recent studies have identified several AMPA-R inter-acting proteins that can modulate the activity and trafficking ofAMPA-Rs (5). In this study, we immunopurified the AMPA-Rfrom rat brain and found that the RhoA GEF, GEF-H1, is asignificant component of the AMPA-R complex in vivo. TruncatedGEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involvedin cell proliferation (6–8). The full-length GEF-H1 was found as a120-kDa protein consisting of 985 aa (9). Interestingly, GEF-H1binding to microtubule regulates its enzymatic activity, suggestingGEF-H1 could mediate cross-talk between microtubules and actin(9, 10). Our functional analysis of GEF-H1 demonstrated thatGEF-H1 negatively regulates the density and length of spinesthrough a RhoA signaling cascade. In addition, AMPA-R activity-dependent changes in spine development were eliminated by inhi-bition of GEF-H1. Together, this study strongly suggests thatGEF-H1 is an AMPA-R associated protein that is important foractivity-dependent structural plasticity.

ResultsIdentification of GEF-H1 as a Component of the AMPA-R Complex.Through proteomic screening of AMPA-R-binding proteins in ratbrains, GEF-H1, a GEF, was identified as a component of theAMPA-R complex in the brain [Fig. 1 and supporting information(SI) Fig. S1]. AMPA-R complexes were purified from rat brainsusing wheat germ agglutinin (WGA)-chromatography and large-scale immunoprecipitation (IP). WGA-chromatography wasadapted to enrich for glycosylated mature membrane proteins andto exclude immature AMPA-R complexes in the endoplasmicreticulum and Golgi apparatus. To distinguish specific binding fromnonspecific binding of proteins to IgG, Protein A, and Sepharoseduring the large-scale IP, the eluant from WGA-chromatographywas divided into 2 pools and 1 pool was used as a negative control.In the negative control group, the GluR1 antibody was blocked withthe synthetic peptide used for the generation of the antibody (seeFig. 1A, Pep �). The isolation of many of the copurifying proteinswas inhibited by preabsorbing the GluR1 antibody with the anti-

Author contributions: M.-G.K. and R.L.H. designed research; M.-G.K. and Y.G. performedresearch; M.-G.K. and Y.G. contributed new reagents/analytic tools; M.-G.K., Y.G., andR.L.H. analyzed data; and M.-G.K. and R.L.H. wrote the paper.

Conflict of interest statement: Under a licensing agreement between Millipore and theJohns Hopkins University, R.L.H. is entitled to a share of royalty received by the universityon sales of products described in this article. R.L.H. is a paid consultant to Millipore. Theterms of this arrangement are being managed by the Johns Hopkins University in accor-dance with its conflict of interest policies.

Freely available online through the PNAS open access option.

1Present address: Department of Neuroscience and Cell Biology, University of Texas MedicalBranch, Galveston, TX 77555.

2To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/cgi/content/full/0812861106/DCSupplemental.

© 2009 by The National Academy of Sciences of the USA

www.pnas.org�cgi�doi�10.1073�pnas.0812861106 PNAS � March 3, 2009 � vol. 106 � no. 9 � 3549–3554

NEU

ROSC

IEN

CE

Dow

nloa

ded

by g

uest

on

May

25,

202

0

Page 2: AMPA receptor and GEF-H1/Lfc complex regulates dendritic ... · GEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involved in cell proliferation (6–8). The full-length GEF-H1

genic peptide, indicting their specific association with AMPA-Rs.As shown in Fig. 1A, this procedure specifically isolated a majorprotein band with a molecular weight of 105 kDa, which containsthe AMPA-R subunits GluR1–4, and several other protein bandsdetected by silver-staining. One of the proteins (see Fig. 1A, arrow)around the 120-kDa region was excised from the protein gel andapplied to liquid chromatography/tandem mass spectrometry (LC-MS/MS) analysis as described in Methods and the SI Methods.Protein database searches identified 3 peptides as a part of Rho/RacGEF 2 (see Fig. S1A). Fig. S1B shows MS/MS spectra of one of thepeptides. Further sequence analysis indicated that the Rho/RacGEF 2 is a rat homolog of human GEF-H1 and mouse Lfcidentified in previous studies (6–9).

The specificity of GEF-H1 association with AMPA-R complexwas confirmed through Western blot analyses with protein samplesfrom rat brains, cultured cortical neurons, and heterologous cells(see Fig. 1 B, C and D). First, protein samples from each step of thepurification of AMPA-Rs were probed with anti-GEF-H1 antibod-ies using Western blot analysis (see Fig. 1B). GEF-H1 was detectedin solubilized brain lysates at around 120 kDa, similar in size to itshuman and mouse homologous. Interestingly, GEF-H1 was de-tected in the eluant of WGA-chromatography as much as in thevoid, suggesting that significant amounts of GEF-H1 bind to maturemembrane proteins. The enrichment and specific detection of theGEF-H1 protein in the eluant of the large-scale AMPA-R IPconfirmed the result of LC-MS/MS analysis (see Fig. 1B). In

small-scale IPs of endogenous AMPA-R complexes using corticalneuronal cultures, GEF-H1 as well as stargazin (a known interactorof AMPA-Rs) was specifically coimmunoprecipitated with GluR1as a part of AMPA-R complex in neurons. Neither GEF-H1 norstargazin was detected in this co-IP when GluR1 antibody wasblocked by the synthetic peptide (see Fig. 1C, Peptide �). Forfurther analysis of GEF-H1 association with AMPA-R complex,GFP-tagged GEF-H1 (GFP-GEF-H1) was coexpressed with myc-tagged AMPA-R subunits in heterologous cells, and the GFP-GEF-H1 was immunoprecipitated using an anti-GFP antibody (seeFig. 1D). Both GluR1 and GluR2 were specifically coimmunopre-cipitated with GFP-GEF-H1. However, the efficiency of this co-IPwith heterologous cell lysate was much lower than that with brainor neuronal lysates, suggesting that the interaction may be stabilizedby other proteins or by posttranslational modifications in neurons.Surprisingly, a GluR1 mutant lacking the cytoplasmic C-terminal(R1Ct�) was also coimmunoprecipitated with GFP-GEF-H1, in-dicating that the C-terminal of AMPA-R is not required forGEF-H1 binding (see Fig. 1D). On the other hand, GluR6 (asubunit of kainate receptors) was not coimmunoprecipitated withGFP-GEF-H1, indicating that the interaction between AMPA-Rsand GEF-H1 is specific (Fig. S2).

GEF-H1 Negatively Regulates the Development of Dendritic Spines.Our biochemical and immunocytochemical analyses of GEF-H1showed localization of GEF-H1 in postsynaptic density (PSD) andspines (Figs. S3A and S4C). In addition, GEF-H1 was highlyexpressed in the brain during the period of active spine develop-ment (Fig. S3B). Given that a GEF-H1 could regulate actindynamics through the activation of Rho GTPases, we hypothesizedthat GEF-H1 could modulate spine development. Because previ-ous studies showed that dendritic spine development starts after 14days in vitro (DIV) in cultured dissociated neurons (11), ourexperiments were designed to manipulate the GEF-H1 activityduring 14 to 16 DIV. We first examined the effects of GEF-H1 lossof function on spine development using a dominant-negativeform of GEF-H1 and shRNA knock-down of GEF-H1 expression.Both of these manipulations demonstrated that GEF-H1 regulatesspine density and length (Figs. 2 and 3). For quantitative analysis inthis study, a spine was defined as a small protrusion from dendritewith GluR1 staining. Therefore, neurons were costained withantibodies recognizing transfected GFP-GEF-H1 and endogenousGluR1. In addition, either GFP or mCherry was used as morpho-logical marker. Through this triple immunofluorescence staining ofcultured hippocampal neurons, the number and length of spinesonly from GFP or GFP-GEF-H1 transfected neurons could bemeasured.

A dominant-negative form of GEF-H1 was generated by intro-ducing a point mutation (T247A) on the catalytic domain (RhoGEF domain) of GEF-H1 (see SI Methods for detail). A group of13 DIV hippocampal neurons was cotransfected with the GFP-tagged dominant-negative GEF-H1 (GEF-DN) and mCherry, fixedat 16 DIV, and stained with antibodies against GFP and GluR1.The neurons transfected with GEF-DN (see Fig. 2 A and A�) havemore spines and longer spines compared with the neurons trans-fected with GFP (see Fig. 2 B and B�). The change on spine densitywas analyzed quantitatively by counting the number of spines per 10�m of dendrite. Over-expression of GEF-DN significantly in-creased spine density (see Fig. 2C and Table S1). The change inspine length was analyzed quantitatively by plotting average andfrequency (%) distribution of spine length. Over-expression ofGEF-DN significantly increased average spine length (see Fig. 2Dand Table S1). The length increase was the result of an increase inpercentage of longer spines (see Fig. 2E). On the other hand,over-expression of GEF-H1 decreased the length of dendriticspines (Fig. 4A and E, and see Table S1). The length decrease isbecause of an increase in the percentage of shorter spines (Fig. 4F)without increase in spine number (see Fig. 4D and Table S1).

A220160120

Pep + -

1208070604030

2010

181.8115.582.2

GEF

B

181.811

Peptide + - + -GEF

48 8

115.5

GluR1115.5

WGA IP-Eluant

C

115.582.2

GluR1

Input IP-Eluant

Stg48.8

D

GluR1115.5

GluR1 - - + - + - - + - + GluR2 - + - - - - + - - -R1Ct∆ - - - + - - - - + -GEF + + + + - + + + + -

GEF

GluR2R1Ct∆

Input IP-Eluant

181.8

115.5

82.2

Fig. 1. ProteomicandWesternblotanalysisofGEF-H1associationwithAMPA-Rcomplex. (A) Silver staining of cortical AMPA-R complex resolved by SDS/PAGE.LC-MS/MS analysis of a band (arrow) around the 120-kDa region revealed GEF-H1(GEF), an interactorofAMPA-R.Westernblotanalyseswithanti-GEF-H1antibodyconfirm the association of GEF-H1 with AMPA-Rs (B–D). The immunoprecipita-tion (IP) was performed with (�) or without (–) peptide block (Pep or Peptide)withthepeptideusedtodeveloptheanti-GluR1antibody (AandC).Westernblotanalyses were performed with protein samples from each step of the AMPA-Rcomplexpurification (B), fromcorticalneuronal culture (C), or fromHEK293Tcells(D). Sol, solubilized brain lysate; Stg, stargazin; WGA, wheat-germ agglutinin. (D)GEF-H1 tagged with GFP (GEF) was coexpressed with myc-tagged GluR1, GluR2,or C-terminal truncated form of GluR1 (R1Ct�). Pull-down of the GEF-H1 withanti-GFP antibodies specifically co-immunoprecipitated these GluRs. Molecular-mass of standards are indicated in kDa on the left (A–D).

3550 � www.pnas.org�cgi�doi�10.1073�pnas.0812861106 Kang et al.

Dow

nloa

ded

by g

uest

on

May

25,

202

0

Page 3: AMPA receptor and GEF-H1/Lfc complex regulates dendritic ... · GEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involved in cell proliferation (6–8). The full-length GEF-H1

To study this in more detail, a lentivirus that expresses GEF-H1shRNA and GFP under the control of 2 promoters was generated,and examined as described in the SI Methods and Fig. S5. First, theknock-down efficiency of the shRNA of GEF-H1 in neurons wasquantitatively analyzed using cortical neuronal culture. Four daysafter viral infection, GFP signals begin to appear and reached aplateau 7 days after the infection. Therefore, 9 DIV hippocampalneurons were infected to knock-down endogenous GEF-H1 ex-pression during initial period for spine development (13 to 16 DIV).The infection of the lentivirus carrying GEF-H1 shRNA knockeddown most of endogenous GEF-H1 expression in cortical neurons(86 � 1.0%, n � 4) (see Fig. 3F). The inhibition of GEF-H1expression also significantly down-regulated GluR1 expression(21 � 3.0%, n � 4) (see Fig. 3F).

For immunocytochemical analysis, hippocampal neurons wereinfected with the lentivirus at 9 DIV, fixed at 16 DIV, and stainedwith antibodies against GFP, GluR1, and MAP2 that were used asmarkers for morphology, the postsynaptic membrane, and dendrite,respectively. Compared with the neurons infected with lentiviruscarrying GFP only (see Fig. 3 B and B�), the neurons infected withlentivirus carrying shRNA of GEF-H1 and GFP (see Fig. 3 A andA�) showed dramatic changes in spine development similar to theneurons transfected with GEF-DN. Inhibition of GEF-H1 expres-sion by shRNA significantly increased the spine density (see Fig. 3C

and Table S1) and spine length (see Fig. 3D and Table S1). Thelength increase was because of an increase in percentage of longerspines (see Fig. 3 C and E), which is consistent with the result ofsimilar analysis with GEF-DN (see Fig. 2). Together, the effects ofdominant-negative GEF-H1 and GEF-H1 shRNA and over-expressed GEF-H1 on spine development clearly showed thatGEF-H1 negatively regulates spine development.

Early studies of GEF-H1 suggested that GEF-H1 could activateboth RhoA and Rac1 (7, 12, 13). However, recent studies ofGEF-H1 homologs consistently demonstrated that GEF-H1 is aRhoA-specific GEF in non-neuronal cells (9, 14–16). To test if

A

BGEF-DN mCherry GluR1 Merge

A’ B’GEF-DN Cont

ine

Num

ber

0.30.4

0.5

0.6Cont GEF-DN

EGFP mCherry GluR1 Merge

Spine Length

% o

f Spi

0.0

0.1

0.2

0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.00.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 more

Spin

e #

/10

μ m

02468

10ContGEF-DN

Spi

ne L

engt

h (μ

m)

0.00.40.81.21.62.0C D

Fig. 2. Dominant-negative form of GEF-H1 increases the density and length ofdendritic spines. (A) Hippocampal neurons (13 DIV) were transfected withmCherry and a dominant-negative form of GEF-H1 tagged with GFP (GEF-DN),andstainedthenwithantibodiesagainstGFPandGluR1at16DIV. (B)Asnegativecontrol, the same batch of neurons were transfected with mCherry and GFP andstained then same way as above. (A� and B�) Images of the whole neuronspresented in (A) and (B), respectively. (Scale bars, 10 �m.) (C) The effect of GEF-DNon spine density was analyzed quantitatively by plotting number (#) of spines per10 �m of dendrite. Compared with GFP-transfected neurons (Cont), the neuronstransfected with GEF-DN had significantly more number of spines. (D) ComparedwithControl,GEF-DNcausedsignificant increase in spine length.The labelofbarsis same as in (C). See Table S1 for values and statistical analysis for (C) and (D). (E)TheeffectofGEF-DNonspine lengthwasalsoanalyzedquantitativelybyplottingfrequency (%) distribution of spine length.

A

BGFP GluR1 MAP2 Merge

B’A’ RNAi Cont

GEF

Vec Ri

Vec Ri

F

GluR1

Neuro-Filament

GEF

ActiveRhoA

Total

G

ber 0.5

0.6Cont RNAi

ESp

ine

# /1

0 µm

02468

10ContRNAi

Spin

e Le

ngth

(µm

)

0.00.40.81.21.62.0C D

RhoA

ActiveRac1TotalRac1

Spine Length

% o

f Spi

ne N

umb

0.0

0.1

0.2

0.30.4

RNAi

0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.00.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 more

Fig. 3. shRNA mediated down-regulation of endogenous GEF-H1 increasesdensity and length of dendritic spines and changes the activity of RhoA andRac1. (A) Hippocampal neurons (9 DIV) were infected with lentivirus carryingshRNA of GEF-H1 and GFP, and stained then with antibodies against GFP,GluR1, and MAP2 at 16 DIV. (B) As negative control, the same batch of neuronswere infected with lentivirus carrying GFP only, and stained then same way asabove. (A� and B�) Image of the whole neurons presented in (A) and (B),respectively. (Scale bars, 10 �m.) (C–E) The density and length of spines wereanalyzed quantitatively as explained in Fig. 2. Compared with neurons in-fected with GFP only (Cont), neurons infected with GFP and shRNA of GEF-H1(RNAi) showed significant increase in spine density and length. See Table S1for values and statistical analysis. (F) Cortical neurons (DIV 9) were infectedwith lentivirus carrying shRNA of GEF-H1 and GFP (Ri) or GFP only (Vec). At DIV16, neurons were harvested and solubilized proteins were then applied toWestern analyses with antibodies against GEF-H1, GluR1, and neurofilament.Infection of the shRNA significantly down-regulated the expression of endog-enous GEF-H1. (G) Cortical neurons were infected and harvested as above andsolubilized proteins were applied to RhoA or Rac1 assay. Compared with theinfection of lentivirus carrying GFP only (Vec), the infection of lentiviruscarrying GFP and GEF-shRNA (Ri) significantly increased and decreased activeRhoA and Rac1, respectively. See Table S2 for values and statistical analysis.

Kang et al. PNAS � March 3, 2009 � vol. 106 � no. 9 � 3551

NEU

ROSC

IEN

CE

Dow

nloa

ded

by g

uest

on

May

25,

202

0

Page 4: AMPA receptor and GEF-H1/Lfc complex regulates dendritic ... · GEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involved in cell proliferation (6–8). The full-length GEF-H1

GEF-H1 could activate either RhoA or Rac1 in neuronal cells, theactivity of RhoA and Rac1 was measured by pulling down activatedRhoA or Rac1 after infection of the lentivirus carrying shRNA ofGEF-H1. In agreement with the recent studies from non-neuronalcells, RhoA activity was significantly decreased by knock-down ofendogenous GEF-H1 (see Fig. 3G and Table S2), indicating that

GEF-H1 is a GEF for RhoA in neurons. On the other hand, Rac1activity was increased by knock-down of GEF-H1 (see Fig. 3G andTable S2). Given that RhoA could inhibit Rac1 activity in neurons(17), the decrease of RhoA activity by the shRNA of GEF-H1 couldresult in the increase of Rac1 activity. This result demonstrated thatGEF-H1 could negatively regulate Rac1 activity by activating RhoAin neurons. Previous studies demonstrated that RhoA could neg-atively regulate spine density and length (18) and that Rac1 couldpositively regulate spine density (11, 19, 20). Therefore, it is likelythat the up-regulation of spine density and length by GEF-H1shRNA is a result of down-regulation of RhoA activity accompa-nying up-regulation of Rac1 activity.

GEF-H1 Regulates the Development of Dendritic Spines Through RhoASignaling Cascade. As mentioned above, data in Fig. 3 imply thatRhoA signaling pathway is involved in the regulation of spinedevelopment by GEF-H1. To test this hypothesis, we attempted tointerfere with the negative regulation of spine development byover-expressed GEF-H1 through pharmacological inhibition of theRhoA signaling pathway. As expected, the inhibition of the RhoAsignaling pathway eliminated the effect of GEF-H1 over-expressionon spine development (see Fig. 4).

Treatment of neurons with a RhoA inhibitor, C3T, significantlyincreased the spine density and length of cultured neurons (see Fig.4 B, D, and E, and Table S1). This result is consistent with previousstudy using brain-slice cultures (18). Moreover, treatment of neu-rons with Y27632, an inhibitor of the kinase ROCK, a downstreameffecter of RhoA, also significantly increased the spine density andlength of cultured neurons (see Fig. 4 C–E, and Table S1). TheY27632 effect on spine length is consistent with a previous studyusing brain-slice culture, although the effect on spine density wasnot observed in this previous study (21). In our experiments, theeffect of Y27632 on spine length was significantly stronger than thatof C3T (see Fig. 4 E and F, and Table S1), which may be becauseof the relatively longer treatment of neurons with Y27632 (3 days)than that of C3T (14–16 h).

Over-expression of GEF-H1 showed a dosage effect on spinedevelopment as well as on dendritic arbor development. Neuronswith large amount of GEF-H1 expression had poor spine devel-opment (see Fig. 4A). However, a neuron with low or mildexpression of GEF-H1 did not show any significant changes in thedevelopment of spines (see Fig. S4C). As mentioned above, aneuron over-expressing GEF-H1 always showed strong colocaliza-tion of GEF-H1 with microtubules (Fig. S4 B and E). Therefore, forour quantification of spine development shown in Fig. 4, only theneurons showing microtubule-like structure of GEF-H1 were se-lected as neurons over-expressing GEF-H1.

The negative effect of GEF-H1 over-expression on spine lengthwas completely eliminated by the inhibitor of RhoA or ROCK (seeFig. 4 B, C, E, and F, and Table S1). The combination of GEF-H1over-expression and inhibition of RhoA-ROCK signaling pathwayclearly demonstrated that GEF-H1 could modulate the spinedevelopment through the RhoA-ROCK signaling pathway (seeFig. 4 and Fig. S6).

The effect of the drugs on the RhoA signaling pathway wasconfirmed through RhoA and Rac1 assays with cortical neurons(see Fig. 4G and Table S2). Most RhoA activity was inhibited byC3T. However, Y27632 did not decrease RhoA activity, confirmingthat ROCK is downstream of RhoA. RhoA activity was slightlyincreased by Y27632, possibly because of negative feedback. Fur-thermore, Rac1 activity was significantly increased by the RhoAinhibitor but not by the ROCK inhibitor. This result is alsoconsistent with the increase of Rac1 activity by shRNA of GEF-H1(see Fig. 3G and Table S2), confirming that Rac1 activity isinhibited by RhoA activity in neurons. All together, these resultsdemonstrated that GEF-H1 could regulate spine developmentthrough the RhoA signaling cascade including ROCK and Rac1.

A

GFP mCherry GFP-GEF mCherry

GEFCont

Cont+C3T GEF+C3TB

GFP mCherry GFP-GEF mCherryCont+Y27632 GEF+Y27632C

G

D

mbe

r

0 50.60.7

Cont GEF

F

E

ActiveRhoA

Cont C3T Y27

Spin

e #

/10

µm

02468

10ContC3TY27

GEF - + - + - +

Spin

e Le

ngth

( µm

)

0.0

0.5

1.0

1.5

2.0

GEF - + - + - +

Spine Length

% o

f Spi

ne N

u m

0.00.10.20.30.40.5 C3T

GEF+C3T Y27 GEF+Y27

0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.00.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 more

TotalRhoAActiveRac1

TotalRac1

Fig. 4. The negative effect of GEF-H1 over-expression on spine length iseliminated by an inhibitor of RhoA or ROCK. (A) Hippocampal neurons (13 DIV)were transfected with mCherry and GFP (Cont) or mCherry and GEF-H1 taggedwithGFP(GEF),andthenstainedwithantibodiesagainstGFPandGluR1at16DIV.(B) Hippocampal neurons were transfected the same way as above, treated withaRhoAinhibitorC3transferase(C3T,1.0�g/ml)overnight(14–16h)at15DIV,andstained as above. (C) Hippocampal neurons were transfected the same way asabove, treated with a ROCK inhibitor (Y27632, 100 �M) for 3 days, and stained asabove. Only GFP or GFP-GEF and mCherry images are shown here for comparison(A–C). (Scale bars, 10 �m.) (D–F) The effect of drug or GEF-H1 on the density andlength of spines were analyzed quantitatively as explained in Fig. 2. Comparedwith untreated neurons (Cont), neurons treated with C3T (C3T) or Y27632 (Y27)showed significant increases in spine density. Spine density did not changed byGEF-H1 over-expression (GEF �). (E) Compared with untransfected neurons (firstbar from the left), neurons over-expressing GEF-H1 (second bar) showed signif-icant decrease in spine length. The change of spine length by GEF-H1 over-expression is eliminated by treatment of C3T (fourth bar) or Y27632 (sixth bar).The drug treatments only (third or fifth bar) significantly increased spine length.The label of bars is the same as in (D). (F) The change of spine length by GEF-H1over-expression was eliminated by the drug treatments. See Table S1 for valuesand statistical analysis (D and E). (G) Cortical neurons were treated with C3T orY27632 as above. At 16 DIV, neurons were harvested and solubilized proteinswere applied to RhoA or Rac1 assay. C3T treatment significantly decreased andincreased the activity of RhoA and Rac1, respectively. See Table S2 for values andstatistical analysis.

3552 � www.pnas.org�cgi�doi�10.1073�pnas.0812861106 Kang et al.

Dow

nloa

ded

by g

uest

on

May

25,

202

0

Page 5: AMPA receptor and GEF-H1/Lfc complex regulates dendritic ... · GEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involved in cell proliferation (6–8). The full-length GEF-H1

GEF-H1 Mediates the AMPA-R Activity-Dependent Regulation of SpineDevelopment. It has been previously reported that AMPA-R ac-tivity regulates the development of dendrites through Rho GTPases(2, 22). Previous studies also demonstrated that AMPA-R activityis involved in the stabilization of spines (2, 22). Blocking AMPA-Ractivity with NBQX significantly reduced spine density (23) andspine motility was inhibited by application of AMPA-R agonists(24). However, the molecular mechanism underlying this regulationis not clear. Our data demonstrating the association of GEF-H1with the AMPA-R complex and the regulation of spine develop-ment by GEF-H1, suggest that GEF-H1 could play a role in theAMPA-R-dependent regulation of spine development. To testthis hypothesis we first examined AMPA-R-dependent regulationof spine development after shRNA knock-down of GEF-H1expression.

As shown previously with organotypic hippocampal slicecultures (23), treatment of cultured neurons with the AMPA-Rantagonist NBQX for 7 days significantly decreased spine den-sity because of elimination of immature spines (Fig. 5 B and E,and see Table S1). Most of the remaining spines after NBQXtreatment are mushroom spines that have a medium length andround head (see Fig. 5 B and E and Table S1). This negativeeffect of NBQX on spine density was completely eliminated bythe down-regulation of GEF-H1 expression (see Fig. 5 C and Eand Table S1). The combination of NBQX treatment anddown-regulation of GEF-H1 demonstrated that GEF-H1 iscritical for the AMPA-R regulation of spine development.

To further examine the effect of NBQX on spine development,the activity of RhoA and Rac1 were measured with or withoutNBQX treatment using cortical neuronal cultures (Fig. 5F and seeTable S2). NBQX treatment significantly increased RhoA activityand decreased Rac1 activity. This activity change of Rho GTPasesby NBQX was completely eliminated by the down-regulation ofGEF-H1 expression (see Fig. 5F and Table S2). Therefore, it islikely that the decrease of spine density by NBQX is a result of anincrease of RhoA activity by activation of GEF-H1. All together,these results strongly suggest that inhibition of AMPA-R activityresults in the activation of GEF-H1 and the modulation of spinedevelopment.

DiscussionRecent studies have implicated AMPA-Rs in the regulation ofactivity-dependent structural plasticity (2). However, the moleculardetails of the intracellular signaling pathways for AMPA-R-dependent structural plasticity are largely unknown. In this study,we have shown that GEF-H1 is associated with the AMPA-Rcomplex and that GEF-H1 is a link from AMPA-R activity changesto the regulation of structural plasticity in neurons. Many numbersof neurological disorders, such as mental retardation, are associatedwith defects in dendritic spine morphology and number of dendriticspines (25). Therefore, our study provides additional insights intothe AMPA-R function and synaptic plasticity in general that maybe relevant to brain development, learning and memory, andneurological disorders.

A previous study of GEF-H1 in neurons showed that GFP-GEF-H1 was localized in the dendritic shaft and translocated tospines only after KCl-dependent depolarization or electrical stim-ulation (26). As mentioned above, our results showed that over-expressed GFP-GEF-H1 was not observed in spines but in dendriticshafts. However, we observed GEF-H1 puncta in spines fromneurons expressing low levels of GFP-GEF-H1 that could be similarto the expression level of endogenous GEF-H1. Our Western blotanalysis showed that endogenous GEF-H1 is highly enriched inPSD III fraction from the brains, strongly suggesting that endog-enous GEF-H1 is in spines where most excitatory synapses arelocated. Our neurons were transfected with GFP-GEF-H1 at DIV13 and examined at DIV 16. In the previous study (26), neuronswere transfected with GFP-GEF-H1 at DIV 7 and 8 and examined

at DIV 16 to 21. This difference in expression time of exogenousGFP-GEF-H1 might be responsible for the different subcellularlocation of GEF-H1. In addition, the previous study (26) used ratGEF-H1 cDNA (encode 958 aa) that is shorter than our ratGEF-H1 cDNA (encode 985 aa). Compared to their shorter cDNA,our full-length cDNA encode 27 aa more in the N-terminal region.Previous studies showed that full-length human GEF-H1 andmouse Lfc have the same size as our GEF-H1 cDNA (9, 15).

Data in this study consistently demonstrated that GEF-H1negatively regulates the development of spines through a RhoAsignaling cascade. Our data also strongly indicate that AMPA-R

A

B

Cont

NBQX

CGFP GluR1 MAP2 Merge

NB+Ri

RNAiD

FES

pine

# /1

0 µ m

0

2

4

6

8ContNBQXNB+RiRNAi

ActiveRhoA

TotalRhoAActiveRac1

TotalRac1

Fig. 5. The negative effect of NBQX on spine density is eliminated by shRNAof GEF-H1. (A) A group of hippocampal neurons (9 DIV) was infected withlentivirus carrying GFP only, then stained with antibodies against GFP, GluR1,and MAP2 at 16 DIV. (B) The same batch of neurons were infected same wayas above and treated with an AMPA-R blocker (NBQX, 20 �M) for 7 days. (C)The same batch of neurons were infected with lentivirus carrying shRNA ofGEF-H1 and GFP and treated with NBQX (20 �M). (D) The same batch ofneurons was infected with lentivirus carrying shRNA of GEF-H1 and GFP. (B–D)Staining was done same way as in (A). (Scale bars, 10 �m.) (E) The effect of drugand shRNA of GEF-H1 on spine density was analyzed quantitatively by plottingnumber (#) of spines per 10 �m of dendrite. Compared with untreatedneurons (Cont,), NBQX treatment (NBQX) significantly decreased spine den-sity. The change of spine density by NBQX was eliminated by infection ofshRNA of GEF-H1 (NB�Ri). (F) Cortical neurons were infected with the lenti-virus (RNAi or Ri) and treated with NBQX (NBQX or NB) as above. At 16 DIV,neurons were harvested and solubilized proteins were applied to the RhoA orRac1 assay. NBQX treatment significantly increased and decreased the activityof RhoA and Rac1, respectively. However, the NBQX effect was eliminated byshRNA of GEF-H1 (NB�Ri). See Table S2 for values and statistical analysis.

Kang et al. PNAS � March 3, 2009 � vol. 106 � no. 9 � 3553

NEU

ROSC

IEN

CE

Dow

nloa

ded

by g

uest

on

May

25,

202

0

Page 6: AMPA receptor and GEF-H1/Lfc complex regulates dendritic ... · GEF-H1/Lfc was first identified as an oncogene (p40/Lfc) involved in cell proliferation (6–8). The full-length GEF-H1

activity negatively regulates GEF-H1 activity, resulting in inhibitionof the synaptic RhoA signaling cascades. These are consistent withthe general conclusion derived from the studies of activity-dependent dendrite arbor growth in vivo using a Xenopus tadpole’svisual system. Glutamate receptor activation by visual stimulationnegatively regulated the RhoA signaling pathway, resulting indendritic arbor growth (2). Furthermore, it has been reported thatAMPA-Rs regulate experience-dependent dendritic arbor growthin vivo (27). Similarly, for spine development, AMPA-R activationby local presynaptic inputs could negatively regulate the RhoA-signaling pathway, resulting in an increase in spine density andstrengthening local connections with the presynaptic inputs.Through similar mechanisms, the RhoA signaling pathway could beactivated when spine development needs to be limited because ofa low level of synaptic inputs.

GEF-H1 is the first GEF identified as a mediator of AMPA-Ractivity-dependent regulation of spine development. AMPA-Rsand Rho GTPases have been known as key players for functionaland structural plasticity, respectively. In that regard, our finding ofGEF-H1 as a linker between these two could give us importantinsight into the molecular mechanisms for synaptic developmentand plasticity. Because structural and functional plasticity seems tobe coordinated, there may be common regulators of functional andstructural plasticity that coordinate these 2 aspects of synapticplasticity. GEF-H1 may be one of the common regulators becauseAMPA-R trafficking is regulated by actin dynamics that is regu-lated by GEF-H1 activity. It has been suggested that synaptic Ca2�

influx through NMDA receptors during long-term potentiationinduction triggers Rho GTPase-meditated actin polymerization,resulting in AMPA-R trafficking to synapses (28). Regulation of theRhoA signaling pathway by GEF-H1 could also be involved in thisNMDA receptor-mediated AMPA-R trafficking in and out ofsynapses.

MethodsDetailed experimental methods are described in SI Methods. The use and careof animals in this study follows the guideline of the Institutional Animal Careand Use Committee at the Johns Hopkins University.

Biochemical Analyses of AMPA-R Complex from Rat Brains and Cells. Prepara-tions of rat brain lysate, fractionation, and solubilization are described in the SIMethods in detail. For the purification of AMPA-R complex from rat brain,matured membrane proteins were then enriched as described in a previous study(29). AMPA-R complexes were then purified through large-scale IP followed byMass Spectrometry analysis as described in the SI Methods and a previous study(30) in detail. Purification of synaptosome and postsynaptic density were per-formed based on a method described previously (31). Western blot analyses wereperformed as described previously (32).

cDNA Subcloning, Mutagenesis, and Preparation of Lentiviral shRNA. The ESTclone of Rho/Rac GEF2 was obtained from Integrated Molecular Analysis ofGenomes and their Expression (IMAGE) Consortium and subcloned into mam-malian-expressionvectors.Basedonapreviousstudy(33), thedominant-negativeform of GEF-H1 (GEF-DN) was designed and generated. The short hairpin RNA(shRNA) of GEF-H1 was designed based on the sequences of short interferenceRNA (siRNA) used in previous studies of GEF-H1 homologous (33, 34), and theshRNA was subcloned into lentiviral vector, FUGW (35). See the SI Methods fordetails.

Neuronal Cell Culture, Immunocytochemistry, Microscopy Image Analysis, andStatistics. Corticalandhippocampalneuroncultureswereprepared,maintained,and analyzed as previously described (36). See SI Methods for details.

ACKNOWLEDGMENTS. We thank Min Dai and Da-ting Lin for technical assis-tance. This work was supported by the National Institutes of Health GrantsR01NS036715 (to R.L.H.) and N01-HV-28180 (to Y.G.). R.L.H. is an investigator ofthe Howard Hughes Medical Institute. M.-G.K. was supported by an EpilepsyFoundation Postdoctoral Fellowship.

1. Dillon C, Goda Y (2005) The actin cytoskeleton: integrating form and function at thesynapse. Annu Rev Neurosci 28:25–55.

2. Van Aelst L, Cline HT (2004) Rho GTPases and activity-dependent dendrite develop-ment. Curr Opin Neurobiol 14:297–304.

3. Jaffe AB, Hall A (2005) Rho GTPases: biochemistry and biology. Annu Rev Cell Dev Biol21:247–269.

4. Tolias KF, et al. (2005) The Rac1-GEF Tiam1 couples the NMDA receptor to theactivity-dependent development of dendritic arbors and spines. Neuron 45:525–538.

5. Shepherd JD, Huganir RL (2006) The cell biology of synaptic plasticity: AMPA receptortrafficking. Annu Rev Cell Dev Biol 23:613–643.

6. Reddy AB, Chatterjee A, Rothblum LI, Black A, Busch H (1989) Isolation and character-ization of complementary DNA to proliferating cell nucleolar antigen P40. Cancer Res49:1763–1767.

7. Ren Y, Li R, Zheng Y, Busch H (1998) Cloning and characterization of GEF-H1, amicrotubule-associated guanine nucleotide exchange factor for Rac and Rho GTPases.J Biol Chem 273:34954–34960.

8. Whitehead I, Kirk H, Tognon C, Trigo-Gonzalez G, Kay R (1995) Expression cloning ofLfc, a novel oncogene with structural similarities to guanine nucleotide exchangefactors and to the regulatory region of protein kinase C. J Biol Chem 270:18388–18395.

9. Krendel M, Zenke FT, Bokoch GM (2002) Nucleotide exchange factor GEF-H1 mediatescross-talk between microtubules and the actin cytoskeleton. Nat Cell Biol 4:294–301.

10. Callow MG, Zozulya S, Gishizky ML, Jallal B, Smeal T (2005) PAK4 mediates morpho-logical changes through the regulation of GEF-H1. J Cell Sci 118:1861–1872.

11. Wiens KM, Lin H, Liao D (2005) Rac1 induces the clustering of AMPA receptors duringspinogenesis. J Neurosci 25:10627–10636.

12. Glaven JA, Whitehead I, Bagrodia S, Kay R, Cerione RA (1999) The Dbl-related protein,Lfc, localizes to microtubules and mediates the activation of Rac signaling pathways incells. J Biol Chem 274:2279–2285.

13. Glaven JA, Whitehead IP, Nomanbhoy T, Kay R, Cerione RA (1996) Lfc and Lsc oncop-roteins represent two new guanine nucleotide exchange factors for the Rho GTP-binding protein. J Biol Chem 271:27374–27381.

14. Aijaz S, D’Atri F, Citi S, Balda MS, Matter K (2005) Binding of GEF-H1 to the tightjunction-associated adaptor cingulin results in inhibition of Rho signaling and G1/Sphase transition. Dev Cell 8:777–786.

15. Benais-Pont G, et al. (2003) Identification of a tight junction-associated guaninenucleotide exchange factor that activates Rho and regulates paracellular permeability.J Cell Biol 160:729–740.

16. Matsuzawa T, Kuwae A, Yoshida S, Sasakawa C, Abe A (2004) EnteropathogenicEscherichia coli activates the RhoA signaling pathway via the stimulation of GEF-H1.EMBO J 23:3570–3582.

17. Li Z, Aizenman CD, Cline HT (2002) Regulation of rho GTPases by crosstalk and neuronalactivity in vivo. Neuron 33:741–750.

18. Tashiro A, Minden A, Yuste R (2000) Regulation of dendritic spine morphology by theRho family of small GTPases: antagonistic roles of Rac and Rho. Cereb Cortex 10:927–938.

19. Nakayama AY, Harms MB, Luo L (2000) Small GTPases Rac and Rho in the maintenanceof dendritic spines and branches in hippocampal pyramidal neurons. J Neurosci20:5329–5338.

20. Penzes P, et al. (2003) Rapid induction of dendritic spine morphogenesis by trans-synaptic ephrinB-EphB receptor activation of the Rho-GEF kalirin. Neuron 37:263–274.

21. Tashiro A, Yuste R (2004) Regulation of dendritic spine motility and stability by Rac1and Rho kinase: evidence for two forms of spine motility. Mol Cell Neurosci 26:429–440.

22. Nimchinsky EA, Sabatini BL, Svoboda K (2002) Structure and function of dendriticspines. Annu Rev Physiol Annu Rev Physiol 64:313–353.

23. McKinney RA, Capogna M, Durr R, Gahwiler BH, Thompson SM (1999) Miniaturesynaptic events maintain dendritic spines via AMPA receptor activation. Nat Neurosci2(1):44–49.

24. Fischer M, Kaech S, Wagner U, Brinkhaus H, Matus A (2000) Glutamate receptorsregulate actin-based plasticity in dendritic spines. Nat Neurosci 3:887–894.

25. Halpain S, Spencer K, Graber S (2005) Dynamics and pathology of dendritic spines. ProgBrain Res 147:29–37.

26. Ryan XP, et al. (2005) The Rho-specific GEF Lfc interacts with neurabin and spinophilinto regulate dendritic spine morphology. Neuron 47(1):85–100.

27. Haas K, Li J, Cline HT (2006) AMPA receptors regulate experience-dependent dendriticarbor growth in vivo. Proc Natl Acad Sci USA 103:12127–12131.

28. Matus A (2005) Growth of dendritic spines: a continuing story. Curr Opin Neurobiol15(1):67–72.

29. Kang MG, et al. (2001) Biochemical and biophysical evidence for gamma 2 subunitassociation with neuronal voltage-activated Ca2� channels. J Biol Chem 276:32917–32924.

30. Guo Y, Ma SF, Grigoryev D, Van Eyk J, Garcia JG (2005) 1-DE MS and 2-D LC-MS analysisof the mouse bronchoalveolar lavage proteome. Proteomics 5:4608–4624.

31. Cho KO, Hunt CA, Kennedy MB (1992) The rat brain postsynaptic density fractioncontains a homolog of the Drosophila discs-large tumor suppressor protein. Neuron9:929–942.

32. Boehm J, et al. (2006) Synaptic incorporation of AMPA receptors during LTP is con-trolled by a PKC phosphorylation site on GluR1. Neuron 51:213–225.

33. Birukova AA, et al. (2006) GEF-H1 is involved in agonist-induced human pulmonaryendothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 290:L540–L548.

34. Bakal CJ, et al. (2005) The Rho GTP exchange factor Lfc promotes spindle assembly inearly mitosis. Proc Natl Acad Sci USA 102:9529–9534.

35. Lois C, Hong EJ, Pease S, Brown EJ, Baltimore D (2002) Germline transmission andtissue-specific expression of transgenes delivered by lentiviral vectors. Science295:868–872.

36. Liao D, Zhang X, O’Brien R, Ehlers MD, Huganir RL (1999) Regulation of morpho-logical postsynaptic silent synapses in developing hippocampal neurons. Nat Neu-rosci 2:37– 43.

3554 � www.pnas.org�cgi�doi�10.1073�pnas.0812861106 Kang et al.

Dow

nloa

ded

by g

uest

on

May

25,

202

0