14
Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 by Cell Press CaMKII b Functions As an F-Actin Targeting Module that Localizes CaMKIIa/ b Heterooligomers to Dendritic Spines while in 10-day-old postnatal mice, the forebrain a:b ratio is 1:1 (Miller and Kennedy, 1985). On a structural basis, recombinant CaMKIIa as well as purified brain CaMKII has been shown to form oligomers with z8 to 12 subunits (Bennett et al., 1983; Kanaseki et al., 1991). Kang Shen, Mary N. Teruel, Kala Subramanian, and Tobias Meyer* Department of Cell Biology Duke University Medical Center Durham, North Carolina 27710 CaMKIIb and CaMKIIa have a similar overall domain organization and corresponding autophosphorylation consensus sequences, and even though the calmodulin Summary binding affinity of CaMKIIb is slightly higher than that of CaMKIIa, the regulation of different CaMKII isoforms Ca 21 /calmodulin-dependent protein kinase II (CaMKII) by Ca 21 /CaM and autophosphorylation is similar overall is a serine/threonine protein kinase that regulates (Miller and Kennedy, 1985; GuptaRoy and Griffith, 1996; long-term potentiation and other forms of neuronal plas- De Koninck and Schulman, 1998). Despite these similari- ticity. Functional differences between the neuronal ties, it has been controversial whether CaMKIIb forms CaMKIIa and CaMKIIb isoforms are not yet known. oligomers on its own (Yamauchi et al., 1989), whether Here, we use green fluorescent protein–tagged (GFP- CaMKIIa and CaMKIIb form heterooligomers when ex- tagged) CaMKII isoforms and show that CaMKIIb is pressed at the same time (Kanaseki et al., 1991), and bound to F-actin in dendritic spines and cell cortex whether the two isoforms are differentially localized while CaMKIIa is largely a cytosolic enzyme. When within cells (Scholz et al., 1988; Nomura et al., 1997). expressed together, the two isoforms form large het- Why is the homo- and heterooligomerization of CaMKIIa erooligomers, and a small fraction of CaMKIIb is suffi- and CaMKIIb important? Earlier studies have shown that cient to dock the predominant CaMKIIa to the actin oligomerization is required for physiological autophos- cytoskeleton. Thus, CaMKIIb functions as a targeting phorylation of a threonine residue at position 286 in module that localizes a much larger number of CaMKII a CaMKIIa (Hanson et al., 1994; Mukherji and Soderling, isozymes to synaptic and cytoskeletal sites of action. 1994). When autophosphorylated at this site (or at threo- nine 287 for CaMKIIb), CaMKII is partially active in the Introduction absence of Ca 21 /CaM (autonomous activity) (Lai et al., 1986; Lou et al., 1986; Miller and Kennedy, 1986; Ca 21 /calmodulin-dependent protein kinase II (CaMKII) Schworer et al., 1986) and exhibits a several hundred– is a ubiquitous kinase that is expressed at high concen- fold increase in calmodulin binding affinity (trapping of trations in neurons and at lower concentrations in most calmodulin) (Meyer et al., 1992). In addition to this regu- other cell types. Previous studies suggested that CaMKII latory function, it is conceivable that oligomerization has is an essential mediator for long-term potentiation and a fundamental role in the localization of CaMKII. Since other forms of synaptic plasticity (reviewed by Soder- CaMKIIa is the predominant isoform in the brain, it can ling, 1993; Braun and Schulman, 1995). Furthermore, be hypothesized that the main function of CaMKIIb iso- forms is to indirectly alter the localization of CaMKIIa CaMKII activity may have an important role in stabilizing isoforms by first forming CaMKIIa/b heterooligomers the dendritic architecture (Wu and Cline, 1998). A critical and then localizing the mixed complex to new docking neuronal function of the a isoform of CaMKII (CaMKIIa) sites. Such a heterologous targeting mechanism would was directly demonstrated by studying mice that were enable cells to alter the cellular localization of CaMKIIa either lacking CaMKIIa or expressed mutated CaMKIIa. indirectly by controlling the ratio of CaMKIIb to CaMKIIa CaMKIIa-deficient mice as well as transgenic mice ex- expression. pressing an autonomously active or an autophosphory- Here, we use green fluorescent protein–tagged (GFP- lation-deficient CaMKIIa showed impaired long-term tagged) CaMKIIa and CaMKIIb isoforms to explore the potentiation as well as defects in spatial learning and subcellular localization and oligomerization of CaMKIIa memory (Silva et al., 1992; Chapman et al., 1995; May- and CaMKIIb. We found that dendritic spines and filo- ford et al., 1995, 1996; Glazewski et al., 1996; Gordon podia as well as the cortical cytoskeleton are the primary et al., 1996; Giese et al., 1998). docking sites for expressed CaMKIIb. In contrast, ex- Since not only CaMKIIa but also CaMKIIb is a promi- pressed CaMKIIa was uniformly distributed in the soma nent isoform in the central nervous system, the question and processes and was largely absent from spines. arises whether CaMKIIa and CaMKIIb have different However, when expressed in the same cell, CaMKIIb roles in regulating neuronal functions. Such functional targeted CaMKIIa to dendritic spines and the cell cortex. differences between the two isoforms would have a di- In vitro binding studies suggested that this targeting rect impact on our understanding of cell type–specific results from a direct binding interaction of CaMKIIb with signaling processes, since the relative expression of F-actin. We then developed a GFP-based protein– CaMKIIa and CaMKIIb is markedly different in different protein interaction assay (“Pull-Out” binding assay) to brain regions and at different developmental stages. For explore the binding interactions between CaMKIIa and example, the ratios of a and b subunits are about 3:1 CaMKIIb isoforms in living cells. When expressed alone, and 1:4 in adult forebrain and cerebellum, respectively, CaMKIIb was found to form homooligomers with an average size that is markedly smaller than the z13 sub- units measured for CaMKIIa homooligomers. When * To whom correspondence should be addressed.

Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 by Cell Press

CaMKIIb Functions As an F-Actin Targeting Modulethat Localizes CaMKIIa/b Heterooligomersto Dendritic Spines

while in 10-day-old postnatal mice, the forebrain a:bratio is 1:1 (Miller and Kennedy, 1985). On a structuralbasis, recombinant CaMKIIa as well as purified brainCaMKII has been shown to form oligomers with z8 to12 subunits (Bennett et al., 1983; Kanaseki et al., 1991).

Kang Shen, Mary N. Teruel, Kala Subramanian,and Tobias Meyer*Department of Cell BiologyDuke University Medical CenterDurham, North Carolina 27710

CaMKIIb and CaMKIIa have a similar overall domainorganization and corresponding autophosphorylationconsensus sequences, and even though the calmodulinSummarybinding affinity of CaMKIIb is slightly higher than thatof CaMKIIa, the regulation of different CaMKII isoformsCa21/calmodulin-dependent protein kinase II (CaMKII)by Ca21/CaM and autophosphorylation is similar overallis a serine/threonine protein kinase that regulates(Miller and Kennedy, 1985; GuptaRoy and Griffith, 1996;long-term potentiation and other forms of neuronal plas-De Koninck and Schulman, 1998). Despite these similari-ticity. Functional differences between the neuronalties, it has been controversial whether CaMKIIb formsCaMKIIa and CaMKIIb isoforms are not yet known.oligomers on its own (Yamauchi et al., 1989), whetherHere, we use green fluorescent protein–tagged (GFP-CaMKIIa and CaMKIIb form heterooligomers when ex-tagged) CaMKII isoforms and show that CaMKIIb ispressed at the same time (Kanaseki et al., 1991), andbound to F-actin in dendritic spines and cell cortexwhether the two isoforms are differentially localizedwhile CaMKIIa is largely a cytosolic enzyme. Whenwithin cells (Scholz et al., 1988; Nomura et al., 1997).

expressed together, the two isoforms form large het-Why is the homo- and heterooligomerizationof CaMKIIa

erooligomers, and a small fraction of CaMKIIb is suffi- and CaMKIIb important? Earlier studies have shown thatcient to dock the predominant CaMKIIa to the actin oligomerization is required for physiological autophos-cytoskeleton. Thus, CaMKIIb functions as a targeting phorylation of a threonine residue at position 286 inmodule that localizes a much larger number of CaMKIIa CaMKIIa (Hanson et al., 1994; Mukherji and Soderling,isozymes to synaptic and cytoskeletal sites of action. 1994). When autophosphorylated at this site (or at threo-

nine 287 for CaMKIIb), CaMKII is partially active in theIntroduction absence of Ca21/CaM (autonomous activity) (Lai et

al., 1986; Lou et al., 1986; Miller and Kennedy, 1986;Ca21/calmodulin-dependent protein kinase II (CaMKII) Schworer et al., 1986) and exhibits a several hundred–is a ubiquitous kinase that is expressed at high concen- fold increase in calmodulin binding affinity (trapping oftrations in neurons and at lower concentrations in most calmodulin) (Meyer et al., 1992). In addition to this regu-othercell types. Previous studiessuggested that CaMKII latory function, it is conceivable that oligomerization hasis an essential mediator for long-term potentiation and a fundamental role in the localization of CaMKII. Sinceother forms of synaptic plasticity (reviewed by Soder- CaMKIIa is the predominant isoform in the brain, it canling, 1993; Braun and Schulman, 1995). Furthermore, be hypothesized that the main function of CaMKIIb iso-

forms is to indirectly alter the localization of CaMKIIaCaMKII activity may have an important role in stabilizingisoforms by first forming CaMKIIa/b heterooligomersthe dendritic architecture (Wu and Cline, 1998). A criticaland then localizing the mixed complex to new dockingneuronal function of the a isoform of CaMKII (CaMKIIa)sites. Such a heterologous targeting mechanism wouldwas directly demonstrated by studying mice that wereenable cells to alter the cellular localization of CaMKIIaeither lacking CaMKIIa or expressed mutated CaMKIIa.indirectly by controlling the ratio of CaMKIIb to CaMKIIaCaMKIIa-deficient mice as well as transgenic mice ex-expression.pressing an autonomously active or an autophosphory-

Here, we use green fluorescent protein–tagged (GFP-lation-deficient CaMKIIa showed impaired long-termtagged) CaMKIIa and CaMKIIb isoforms to explore thepotentiation as well as defects in spatial learning andsubcellular localization and oligomerization of CaMKIIamemory (Silva et al., 1992; Chapman et al., 1995; May-and CaMKIIb. We found that dendritic spines and filo-ford et al., 1995, 1996; Glazewski et al., 1996; Gordonpodia as well as the corticalcytoskeleton are the primaryet al., 1996; Giese et al., 1998).docking sites for expressed CaMKIIb. In contrast, ex-Since not only CaMKIIa but also CaMKIIb is a promi-pressed CaMKIIa was uniformly distributed in the somanent isoform in the central nervous system, the questionand processes and was largely absent from spines.arises whether CaMKIIa and CaMKIIb have differentHowever, when expressed in the same cell, CaMKIIbroles in regulating neuronal functions. Such functionaltargeted CaMKIIa todendritic spines and the cell cortex.

differences between the two isoforms would have a di-In vitro binding studies suggested that this targeting

rect impact on our understanding of cell type–specificresults from a direct binding interaction of CaMKIIb with

signaling processes, since the relative expression of F-actin. We then developed a GFP-based protein–CaMKIIa and CaMKIIb is markedly different in different protein interaction assay (“Pull-Out” binding assay) tobrain regions and at different developmental stages. For explore the binding interactions between CaMKIIa andexample, the ratios of a and b subunits are about 3:1 CaMKIIb isoforms in living cells. When expressed alone,and 1:4 in adult forebrain and cerebellum, respectively, CaMKIIb was found to form homooligomers with an

average size that is markedly smaller than the z13 sub-units measured for CaMKIIa homooligomers. When*To whom correspondence should be addressed.

Page 2: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

Neuron594

expressed at the same time, CaMKIIb isoforms incor-porated equally well into either CaMKIIa or CaMKIIboligomers (and vice versa). Half-maximal targeting ofCaMKIIa oligomers to the cytoskeleton was achieved ifat least 15% of CaMKIIb were present in the same cell,suggesting that a small number of CaMKIIb subunitsare required to dock CaMKIIa/b heterooligomers withz13 subunits to F-actin. Our studies suggest that thesynaptic localization of CaMKII activity is controlledby the relative expression of CaMKIIb F-actin–dockingmodules.

Results

Expressed CaMKIIb but Not CaMKIIa Is Enrichedin Dendritic Branches and Cell CortexWe investigated the cellular localization of CaMKIIa ver-sus CaMKIIb isoforms by constructing CaMKII fusionproteins with GFP (Figure 1A). An earlier study has shownthat a GFP–CaMKIIa construct can phosphorylate sub-strate peptides as well as autophosphorylate itself atthreonine 286 (Shen and Meyer, 1998). An additionalcriterion for functionally intact GFP–CaMKII is the pres-ervation of secondary calcium-independent autophos-phorylations at the sites that prevent further calmodulinbinding (residues threonine 305 or 306) (Colbran andSoderling, 1990; Hanson and Schulman, 1992). Wetested whether the GFP tag on CaMKIIa or CaMKIIbaffects either type of autophosphorylation and deter-mined whether the extent of autophosphorylation issim-ilar to that of the wild-type enzyme. Indeed, wild-typeCaMKIIa and GFP-tagged CaMKIIa and CaMKIIb iso-forms showed similar amounts of calcium-dependentand -independent autophosphorylation activity (Figures1B and 1C).

When GFP-tagged CaMKIIa or CaMKIIb isoforms wereexpressed in cultured hippocampal neurons, CaMKIIawas largely homogeneous in the soma and main pro-cesses (Figure 1D, left) but was only minimally presentin the finer branch structures. In contrast, CaMKIIbshowed a striking enrichment in dendritic branches aswell as at the cell cortex (Figure 1D, right). When ex-pressed in rat basophilic leukemia (RBL) cells, CaMKIIawas nearly homogeneously distributed in the cytosoland CaMKIIb had a distinct cortical localization (Figure1E, right).The differential distribution of the two isoformssuggests that CaMKIIb has specific binding interactionsin cells that do not occur for CaMKIIa.

poration and [35S]Met incorporation). The dark bars show CaMKIIFigure 1. Cellular Localization and Mobility of GFP-Tagged CaMKIIa autophosphorylation after incubation with [32P]ATP in high Ca21/and CaMKIIb CaM for 30 s. The light bars show the calcium-independent “burst”(A) Schematic representation of the domain organization of the GFP- autophosphorylation after 30 s in high calcium and 120 s in EGTA.tagged CaMKII isoforms. The catalytic domain (C), regulatory do- (D) Confocal image of GFP-tagged CaMKIIa (left) andCaMKIIb (right)main (R), variable domain (V), and oligomerization domain (A) are expressed in living hippocampal CA1–CA3 neurons. Notice the ho-shown. mogeneous distribution of GFP–CaMKIIa throughout the soma and(B) Autophosphorylation of GFP-tagged CaMKII isoforms. Compari- major branches, whileCaMKIIb is enriched in the dendritic branches.son of the baseline (left), calcium/CaM-dependent (middle), and burst (E) Confocal imageof GFP-tagged CaMKIIa (left) and CaMKIIb (right)(right) autophosphorylation activity of CaMKIIa, GFP–CaMKIIa, and expressed in living RBL cells. Notice the cortical staining and non-GFP–CaMKIIb. The kinase activity of the in vitro translated con- uniform internal staining of GFP–CaMKIIb and the homogeneousstructs are shown. Translated GFP alone was included as a control. distribution of GFP–CaMKIIa.(C) Relative kinase activity corrected for the amount of expressed (F) Comparison of the calculated diffusion coefficients for GFP–CaMKII or GFP–CaMKII protein (measured as the ratio of 32P incor- CaMKIIa and GFP–CaMKIIb.

Page 3: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

CaMKII Targeting595

Figure 2. CaMKIIb Localizes to DendriticSpines and Filopodia in Neurons

(A) Colocalization of GFP–CaMKIIb with thePSD marker PSD-95. Cells were fixed andstainedwith anti–PSD-95 antibodies. The dis-tribution of GFP–CaMKIIb (green) was thencompared to a Cy3-labeled secondary anti-body (red) to visualize the distribution of PSD-95. Arrows point to dendritic spines enrichedin PSD-95 and GFP–CaMKIIb.(B) Magnified region of (A).(C) In some neurons, a filopodia-like stainingby GFP–CaMKIIb can be observed that isreminiscent of developing dendritic spines(left). At right, a magnified view of such filo-podia is shown.(D) Comparison of the localization of GFP–CaMKIIb and active presynaptic terminals inliving neurons.(Left) GFP–CaMKIIb fluorescence (green).(Center) Subtracted image of presynaptic ter-minals loaded with FM 4-64 (red).(Right) Overlay of GFP–CaMKIIb (green) andactive presynaptic terminals (red). Arrowspoint to locations where the presynapticmarker FM 4-64 is juxtaposed to the localizedregions of GFP–CaMKIIb.(E) Control measurement, showing the re-spective distributions of GFP–CaMKIIa (green)and active presynaptic terminals (red) as wellas their overlay.

We tested more directly whether CaMKIIb has more the GFP–CaMKIIb fluorescence recovered on the timescale of 15 s after the laser bleach pulse. Together,binding interactions than CaMKIIa by comparing the

local fluorescence recovery after photobleaching of these measurements suggest that CaMKIIa expressedalone is a highly mobile protein that has only limitedGFP–CaMKIIb to that of GFP–CaMKIIa. A 2 mm diameter

laser photobleach spot was generated in the cell by a cytosolic binding interactions, while CaMKIIb is boundina reversible manner to dendritic and cortical structures.short laser pulse and the fluorescence recovery was

monitored by rapid confocal imaging. Consistent withthe hypothesis that CaMKIIb but not CaMKIIa under- CaMKIIb Is an F-Actin–Docking Module

Enriched in Dendritic Spinesgoes binding interactions, the recovery after photo-bleaching was significantly more rapid for CaMKIIa What are the structures in the dendritic branches and

cell cortex targeted by CaMKIIb? The markedly punctatecompared to that for CaMKIIb. This could be quantita-tively shown by a calculated average diffusion coeffi- staining suggested that CaMKIIb is enriched in dendritic

spines. Indeed, an antibody against the postsynapticcient of CaMKIIb that was five times lower than that ofCaMKIIa (Figure 1F; see Experimental Procedures for a protein PSD-95 showed a clear colocalization between

GFP–CaMKIIb (Figure 2A, green) and PSD-95 (red). Thedescription of the analysis). Nevertheless, the bindinginteractions of CaMKIIb were reversible, since most of arrows in the right panel point to presumed dendritic

Page 4: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

Neuron596

spines, which were highly enriched in PSD-95 and GFP–CaMKIIb (expressed by the yellow color in the overlayimage). A magnification of the dendritic region is shownin Figure 2B. Some of the neurons also showed an en-riched staining of GFP–CaMKIIb in filopodia-like branches(Figure 2C) that were reminiscent of developing dendriticspines (Ziv and Smith, 1996). A magnified image of anarbor of such filopodia is shown in the right panel.

We verified that the dendritic spines marked by anti–PSD-95 antibodies and GFP–CaMKIIb corresponded tomature postsynaptic terminals by comparing the distri-bution of GFP–CaMKIIb to that of functional presynapticterminals. The terminals were marked with the fluores-cent synaptic vesicle marker FM 4-64 using a doubledepolarization protocol (Ziv and Smith,1996; see Experi-mental Procedures). Since this type of colocalizationstudy can be performed in living neurons, potential dis-tribution artifacts that may arise during the fixation ofneurons can be excluded. When comparing the distribu-tion of GFP–CaMKIIb (Figure 2D, green) to the locationof active presynaptic terminals (represented in red as adifference image of the released FM 4-64 after depolar-ization), the overlayed image showed a marked juxta-posed localization of GFP–CaMKIIb and loaded FM 4-64(Figure 2D, right). This suggests that CaMKIIb is indeedenriched in mature dendritic spines. In contrast, the uni-formly distributed GFP–CaMKIIa was not enriched nearactive synapses (Figure 2E).

Since actin is highly enriched in dendritic spines andcell cortex (Caceres el al., 1983; Landis and Reese, 1983;Fisher et al., 1998), it is conceivable that the localizationof CaMKIIb to dendritic spines is mediated by a director indirect binding interaction of CaMKIIb with F-actin.We tested the possible colocalization of CaMKIIb andF-actin in two model cell lines by using rhodamine phal- Figure 3. CaMKIIb Colocalizes with the Cortical Actin Cytoskeletonloidin as a marker for polymerized actin (F-actin). In RBL and Actin Stress Fibers by Binding to F-Actincells, which have predominant cortical F-actin struc- (A) Colocalization of GFP–CaMKIIb (left, green) with rhodamine phal-tures, the cortical rhodamine phalloidin closely colocal- loidin (center, red) in fixed RBL cells. The significant fraction ofized with GFP–CaMKIIb (Figure 3A, GFP–CaMKIIb in yellow structures observed in the overlay image (right) is suggestive

of a nearly complete colocalization of CaMKIIb with polymerizedgreen and rhodamine phalloidin in red). The same nearlyactin.complete overlap was also observed in NIH-3T3 cells,(B) Colocalization of GFP–CaMKIIb (left, green) with rhodamine phal-which are rich in actin stress fibers (Figure 3B). Again,loidin (center, red) in fixed NIH-3T3 cells. Again, a nearly complete

GFP–CaMKIIb is shown in green (left) and phalloidin in colocalization is apparent from the yellow color of stress fibersred (middle), and the overlay image is shown in the (right).right panel. This suggests that the actin cytoskeleton (C) The cortical distribution of GFP–CaMKIIb in RBL cells is dis-colocalization of CaMKIIb is not cell type specific. rupted by latrunculin (top) (50 mM, 2 min). The stress fiber staining

is disrupted by latrunculin (bottom) (50 mM, 2 min). Calibration bars,Further support for the colocalization of CaMKIIb with5 mm.F-actin in living cells was obtained by comparing cells(D) CaMKIIb directly binds to purified F-actin. Nonmuscle G-actinbefore and after treatment with the actin depolarizingwas polymerized at concentration of 150 mg/ml (left). In vitro trans-

drug latrunculin (Spector et al., 1989) (Figure 3C). In RBL lated CaMKII isoforms were added and incubated on ice for 1 hrcells (top) and fibroblasts (bottom), addition of latrun- followed by centrifugation at 100,000 3 g for 30 min. The pelletculin led to a nearly complete loss in the cortical as well and supernatant were resolved by SDS-PAGE and exposed on aas stress fiber staining of GFP–CaMKIIb. Phosphorimager. CaMKIIb is significantly enriched in the actin pellet

when compared to CaMKIIa (right) (average of five experiments).We then determined biochemically whether expressedCaMKIIb can bind directly to purified F-actin. Indeed,Met-35S-labeled CaMKIIb could be effectively sedimented

CaMKIIa Is Targeted to Dendritic Spinesby polymerized actin (Figures 3D, left). In contrast,When Coexpressed with CaMKIIbCaMKIIa is much less sedimentable by polymerized ac-We then tested whether the coexpression of CaMKIIatin using the same assay. Figure 3D, right, shows theand CaMKIIb in the same cell affects their respectivecounts in each of thebands using a bardiagram (averagelocalization. An effective coexpression of both iso-of five measurements). This suggests that the colocali-forms was made possible by using an RNA transfectionzation of CaMKIIb with the actin cytoskeleton observedmethod. In this approach, a large number of translation-in living cells is the result of a direct and reversible

binding interaction between CaMKIIb and F-actin. competent RNA molecules are directly introduced into

Page 5: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

CaMKII Targeting597

Figure 4. CaMKIIa Is Targeted to the ActinCytoskeleton When Expressed in the SameCell with CaMKIIb

(A) Coexpression of GFP–CaMKIIa togetherwith CaMKIIb without a GFPtag in a CA1–CA3neuron.(B) Coexpression of GFP–CaMKIIa togetherwith CaMKIIb without a GFP tag in RBL cells.(C) Comparison of the localization of GFP–CaMKIIa, expressed together with CaMKIIb,to the PSD marker PSD-95. Cells were fixedandstained with anti–PSD-95 antibodies. Thedistribution of GFP–CaMKIIa (green) wascompared to a Cy3-labeled secondary anti-body (red) to visualize the distribution of PSD-95. Arrows point to dendritic spines enrichedin PSD-95 and GFP–CaMKIIa.(D) Localization of GFP–CaMKIIa (mixed withCaMKIIb RNA) compared with active presyn-aptic terminals in living neurons.(Left) GFP–CaMKIIa fluorescence (green).(Center) Subtracted FM 4-64 staining as de-scribed in Figure 2.(Right) Overlay of the two images. Arrowshighlight regions whereGFP–CaMKIIa (mixedwith CaMKIIb RNA) and active presynapticterminals are juxtaposed to each other. Cali-bration bars, 10 mm.(E) Western blots of supernatant and actinpellet fractions of NIH-3T3 cells expressingCaMKIIa, CaMKIIb, or CaMKIIa 1 CaMKIIb,respectively.(Left) An anti-CaMKIIa antibody identifiedmost of the expressed CaMKIIa in the super-natant andonly a minimal amount inthe actin-enriched pellet (see Experimental Proceduressection).(Center) An anti-CaMKIIb antibody identifieda significant fraction of expressed CaMKIIbin the actin pellet.(Right) In cells coexpressing CaMKIIa andCaMKIIb, an anti-CaMKIIa antibody identi-fied a significant fraction of CaMKIIa in theactin pellet, supporting the hypothesis thatCaMKIIb targets CaMKIIa to F-actin.

the cytosol of adherent cells by microporation (Yokoe be confirmed biochemically? We used a detergent ex-traction procedure of NIH-3T3 cells to test whetherand Meyer, 1996; Teruel and Meyer, 1997). Thus, RNA

encoding different proteins can be mixed and expressed CaMKIIb and/or CaMKIIa could be found in an actin-enriched pellet fraction. This extraction protocol hasat a defined ratio within each transfected cell. Strik-

ingly, when GFP–CaMKIIa was expressed together with been shown to significantly enrich for actin and actinbindingproteins (Egelhoffet al., 1991). Expressed CaMKIIbCaMKIIb (without a GFP tag) in hippocampal neurons,

GFP–CaMKIIa became associated with the same den- and CaMKIIa isoforms without a GFP tag were used forthese measurements. In agreement with the in vivo data,dritic spine and cortical structures (Figure 4A). A largely

cortical localization of GFP–CaMKIIa was also observed CaMKIIa, when expressed alone, was largely absentfrom the actin pellet, while CaMKIIb was highly enrichedin RBL cells in the presence of CaMKIIb (Figure 4B). In

contrast, expression of CaMKIIa (without a GFP tag) in the actin pellet (Figure 4E, left and center). When bothisoforms were expressed together, a significant fractiontogether with a similar amount of GFP–CaMKIIb did not

affect the cortical localization of GFP–CaMKIIb (data of CaMKIIa was found in the actin cytoskeletal fraction(Figure 4E, right). Taken together, these studies are con-not shown). Using the same colocalization protocols as

described before in Figure 2, we also found a marked sistent with a targeting mechanism by which CaMKIIais localized to F-actin if expressed together with a suffi-colocalization between GFP–CaMKIIa, coexpressed with

CaMKIIb, and anti PSD-95 antibodies (Figure 4C). In cient amount of CaMKIIb.How does CaMKIIb target CaMKIIa to the actin cy-living neurons,GFP–CaMKIIa, coexpressed with CaMKIIb,

showed a marked localization juxtaposed to the presyn- toskeleton? A likely hypothesis is that CaMKIIa does notundergo cytoskeletal binding interactions of its own butaptic marker FM 4-64 (Figure 4D).

Can this effect of CaMKIIb on CaMKIIa localization binds to CaMKIIb, which then anchors the complex to

Page 6: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

Neuron598

Figure 5. Development of a Pull-Out BindingAssay to Study Protein–Protein Interactionsin Living Cells

(A) Principle of the Pull-Out binding assay.The binding interaction between a Protein Xand Protein Y can be measured by taggingProtein X with an inducible plasma membranebinding domain (PM domain) and Protein Ywith GFP. If a significant fraction of the twoproteins bind to each other, drug addition tar-gets the GFP to the plasma membrane. Incontrast, the cytosolic distribution remainsunaltered if Protein X and Y do not bind toeach other.(B) Property of a minimal phorbol ester bind-ing domain used as an inducible PM domainin the Pull-Out binding assay. A fusion proteinbetween GFP and the phorbol ester bindingdomain can be pulled from the cytosol to theplasma membrane by addition of phorbolester.(Left) Distribution of the fusion protein beforephorbol ester addition.(Right) Distribution of the fusion protein afterphorbol ester addition.(Bottom) Line scans of the fluorescence in-tensity across the cell before and after phor-bol ester addition.(C) Demonstration that nearly all CaMKIIamolecules are part of oligomers. Phorbol es-ter addition to cells with coexpressed PM–CaMKIIa and GFP–CaMKIIa leads to the nearcomplete plasma membrane translocation ofGFP–CaMKIIa.(D) Control measurements showing that ex-pressed GFP itself is not affected by phorbolester addition. Calibration bars, 10 mm.

F-actin. To understand this heterologous CaMKII tar- investigated Protein X and Protein Y could then be inves-tigated by fusing this PM domain to Protein X and ageting mechanism, several important questions about

the binding interaction and oligomerization of CaMKIIa GFP tag to Protein Y (or vice versa). Both fusion proteinscould then be expressed in the same cell and their bind-and CaMKIIb have to be answered. First, can CaMKIIb

form oligomers on its own and, if it does, how large are ing interaction investigated. First, if the initially cytosolicGFP–Protein Y remains cytosolic after drug addition, nothese oligomers compared to those formed by CaMKIIa?

Second, do coexpressed CaMKIIa and CaMKIIb iso- significant binding interaction occurs between ProteinsX and Y. Second, if the drug addition leads to the GFP–forms assemble into hetero- or homooligomers in living

cells? Third, if they form heterooligomers, does the in- Protein Y translocation to the plasma membrane (alongwith the nonfluorescent PM–Protein X), Proteins X andcorporation of CaMKIIb into CaMKIIa oligomers occur

as a stochastic process? Fourth, what minimal ratio of Y bind to each other.We used the first phorbol ester binding domain ofCaMKIIb to CaMKIIa is required for targeting CaMKIIa

to the actin cytoskeleton? protein kinase C as such a PM domain. This small 6 kDadomain is an initially cytosolic protein that binds nearlyirreversibly to the plasma membrane after phorbol esterOligomer Formation Can Be Explored in Living Cellsaddition (Oancea et al., 1998). The distinct property ofUsing a GFP-Based Pull-Out Binding Assaythis domain is shown in Figure 5B. Before phorbol esterTo address the question of homo- versus heterooligomeraddition, a fusion protein of the phorbol ester bindingformation, we developed an assay to quantitativelydomain with GFP is a cytosolic protein (left) that isstudy protein–protein binding interactions in living cells“pulled” from the cytosol to the plasma membrane after(Pull-Out binding assay; Figure 5A). Our strategy was toaddition of phorbol ester (right). This translocation pro-use a protein domain (PM domain) that translocates tocess occurs in less than a minute and is mediated by athe plasma membrane in response to the addition of

a drug. The potential binding interaction between the diffusion-dependent high affinity binding interaction of

Page 7: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

CaMKII Targeting599

the fusion protein with plasma membrane–localized by the average cytosolic fluorescence intensity beforephorbol ester addition (Ipre) (Figure 6E). A DPM:Ipre ratiophorbol ester (Oancea et al., 1998). It should be noted

that the also visible nuclear-localized GFP fusion protein of 0 indicates that no plasma membrane translocationoccurred, and a PM:Ipre ratio of z3 corresponds to thetranslocates to theplasma membrane much more slowly

due to its slow diffusion through nuclear pores. translocation observed for the PM–GFP construct itself.Based on this analysis, a titration curve can be ob-We first used this PM domain to determine whether

most of the expressed CaMKIIa isoforms is present in tained, showing the relative plasma membrane translo-cation at decreasing dilution ratios of PM–CaMKIIa toan oligomeric state. To test for oligomerization in vivo,

PM-tagged CaMKIIa and GFP–CaMKIIa were expressed GFP–CaMKIIa (Figure 6F). A 50% reduction in plasmamembrane targeting was observed at an approximatein the same cell (Figure 5C, left). As expected, addition

of phorbol ester led to a marked translocation of the dilution ratio of 1 PM–CaMKIIa per 14 GFP–CaMKIIa. APoisson distribution model would predict that the proba-initially cytosolic GFP–CaMKIIa to the plasma mem-

brane (Figure 5C, right). This change in the distribution bility (p) to introduce at least one subunit into an oligo-mer with N subunits is p 5 1 - (R/[R 1 1])N, with R asbefore and after addition of phorbol ester can be more

quantitatively measured in a line profile analysis com- the dilution ratio. Using this model, a best fit to the datawas obtained for N 5 13.5, in close agreement with aparing the fluorescence intensity across the cell before

and after phorbol ester addition (Figure 5C, bottom). In previous estimate of 12 subunits for purified CaMKIIa(Yamauchi et al., 1989; Kanaseki et al., 1991).control experiments, GFP alone was coexpressed with

PM-tagged CaMKIIa, and no plasma membrane translo- We then used the same approach to determine ifCaMKIIb forms oligomers. Although GFP–CaMKIIb hascation of GFP was observed after phorbol ester addition

(Figure 5D). The finding that most GFP–CaMKIIa is pulled a partial cortical and internal F-actin localization, thisbinding interaction was reversible, and a much moreto the plasma membrane by PM–CaMKIIa strongly sug-

gests that most of the expressed CaMKIIa is present in pronounced plasma membrane localization can be in-duced by addition of phorbol ester to a PM-taggedthe cell in an oligomeric state.CaMKIIb. Using this phorbol ester–triggered increasein plasma membrane translocation of GFP–CaMKIIb, a

CaMKIIb Homooligomers Are Significantly titration curve was obtained for the relative plasmaSmaller Than CaMKIIa Homooligomers membrane translocation at decreasing dilution ratios ofWhile CaMKIIa has been proposed to be an homooligo- PM–CaMKIIb to GFP–CaMKIIb. Interestingly, the appar-mer with 8–12 subunits, it is controversial if CaMKIIb ent average size of CaMKIIb oligomers was 4.2, signifi-forms oligomers and how large these potential oligo- cantly smaller than that of the a isoform (Figure 6F).mers are (Yamauchi et al., 1989). To measure the appar- Thus, these in vivo measurements clearly show thatent oligomer sizes of CaMKIIa and CaMKIIb in living CaMKIIb can form oligomers, albeit with a significantlycells, we expressed GFP–CaMKII together with a de- smaller apparent size than those formed by CaMKIIa.creasing amount of PM–CaMKII and measured the phor- This apparent size of CaMKIIb oligomers was consistentbol ester–induced translocation to the plasma mem- with our finding that CaMKIIb purified from baculovirus–brane (a schematic view of the expected oligomer transfected Sf9 cells had a size much smaller than thattranslocation process is shown in Figure 6A). Since the of CaMKIIa expressed by the same method (K. S., un-phorbol ester-induced plasma membrane affinity of the published data).PM domain is nearly irreversible, it is likely that a singlesubunit of PM–CaMKIIa is sufficient to induce theplasma membrane translocation of a CaMKIIa oligomer. CaMKIIa and CaMKIIb Form

Stochastic HeterooligomersAs discussed above, the RNA transfection methodallows one to quantitatively titrate the amount of the Since the relative expression of CaMKIIa to CaMKIIb is

highly variable between different types of neurons, wetwo CaMKII fusion proteins in the same cell. To deter-mine the respective expression levels for the two micro- determined how efficient heterooligomer formation is

compared with homooligomer formation. We pursuedporated RNA species, the concentration of the trans-lated proteins was measured in parallel by in vitro the same dilution approach as described in Figure 6

but for the heterooligomers. The calculated line plots intranslation of the same RNAs in the presence of [35S]Met(Figure 6B). Figure 7A show the curves expected for the insertion of

PM–CaMKIIb into GFP–CaMKIIa oligomers for a sto-Figure 6C shows the GFP–CaMKIIa plasma membranetranslocation at increasing dilutions of coexpressed chastic insertion mechanism (dashed curve). The mea-

sured relative plasma membrane translocation for de-PM–CaMKIIa. The left images show the distribution be-fore and the right images after phorbol ester addition. creasing ratios of PM–CaMKIIb to GFP–CaMKIIa closely

matched a predicted stochastic insertion mechanism.Interestingly, the phorbol ester–induced targeting to theplasma membrane was still measurable when PM– The same measurements were then made for PM–

CaMKIIa insertion into GFP–CaMKIIb oligomers by dilu-CaMKIIa was diluted to ,3% of GFP–CaMKIIa. The se-quential reduction in plasma membrane translocation tion of the PM–CaMKIIa fusion protein at a constant

concentration of CaMKIIb (Figure 7B). The calculatedcan be seen more clearly in fluorescence line intensitytraces (Figure 6D). The loss in phorbol ester–mediated curve for a stochastic insertion mechanism is overlap-

ping with the fitted one.plasma membrane targeting at increasing PM–CaMKIIadilutions was analyzed by dividing the relative plasma Together, this titration approach suggests that if

CaMKIIa and CaMKIIb isoforms are expressed at themembrane fluorescence intensity of GFP–CaMKIIa (DPM)

Page 8: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

Neuron600

Figure 6. CaMKIIa Forms Larger OligomersThan CaMKIIb

(A) Schematic representation of the assay tomeasure the size of CaMKIIa and CaMKIIboligomers in living cells.(B) Quantitative comparison of theconcentra-tions of expressed GFP–CaMKIIa and PM–CaMKIIa measured by invitro translation. Therelative concentration of the expressed pro-teins was compared by [35S]Met incorporationinto in vitro translated proteins. The sameRNA was used for the in vitro translation andthe RNA transfection of cells.(C) Only for dilutions below 10% does PM–CaMKIIa begin to lose its potency to translo-cate GFP–CaMKIIa to the plasma membrane.Dilutions were achieved by mixing the RNAsfor PM–CaMKIIa and GFP–CaMKIIa at de-creasing ratios.(Top and center) Plasma membrane translo-cation is still near maximal at dilutions of 1:1and 1:5.(Bottom) Plasma membrane translocation ismarkedly reduced at a 1:40 dilution. Calibra-tion bars, 10 mm.(D) Line scan profiles of three different dilu-tions after phorbol ester addition.(E) Schematic representations of the quanti-tative analysis used to measure the relativeplasma membrane translocation. Ipre and PMare measured before and after PMA addition,respectively.(F) Plot of the relative plasma membranetranslocation of CaMKIIa and CaMKIIb at de-creasing ratios of expressed PM–CaMKII andGFP–CaMKII. Each point is an average of atleast 10 experiments. The solid curves arebest fits to the two set of data, and the dashedlines show the confidence interval. Best fitswere obtained assuming an average of 13.5subunits for CaMKIIa and 4.2 subunits forCaMKIIb.

same time and place, they form mixed oligomers with not resolve whether individual or multiple CaMKIIb sub-a stochastic probability for the insertion of either one units are required for the targeting of CaMKIIa/b hetero-of the isoforms. This also suggests that most CaMKII oligomers to the actin cytoskeleton. We used the sameoligomers in neurons contain a variable fraction of RNA dilution strategy to determine at which ratio ofCaMKIIb that is defined by the relative expression level GFP–CaMKIIa to CaMKIIb the cytoskeletal localizationof locally translated CaMKIIb versus CaMKIIa. For the still occurs. The distinct cortical actin cytoskeleton lo-physiological situation, it is then important to know how calization of CaMKIIb in RBL cells was used in this assaymany CaMKIIb isoforms have to be inserted into mostly (Figure 7C). While CaMKIIb was less potent in targetingCaMKIIa heterooligomers to still effectively target CaMKII GFP–CaMKIIa to the plasma membrane than the PM–to its cytoskeletal docking site. CaMKII constructs, 50% translocation to the cortical

actin cytoskeleton required a ratio of z6.5:1 of GFP–CaMKIIa to CaMKIIb. Since CaMKIIa isoforms containA Small Number of CaMKIIb Isoforms Are Sufficientz13 subunits, this suggests that a small number ofto Target CaMKII HeterooligomersCaMKIIb subunits are sufficient to target CaMKIIa/b het-to the Actin Cytoskeletonerooligomers to the actin cytoskeleton.While the previous studies were useful to dissect the

oligomer formation of CaMKIIa and CaMKIIb, they did In a second independent approach to understand the

Page 9: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

CaMKII Targeting601

Figure 7. Requirement for More Than One CaMKIIb Subunit for Targeting CaMKIIa/b Heterooligomers to the Actin Cytoskeleton

(A) Insertion of CaMKIIb into heterooligomers of mostly CaMKIIa is a stochastic process. The panel shows a plot of the relative plasmamembrane translocation of GFP–CaMKIIa at decreasing ratios of expressed PM–CaMKIIb. Each point is an average of at least ten experiments.The dashed line shows the predicted curve for a stochastic insertion of PM–CaMKIIb into heterooligomers (p 5 1 2 [R/(R 1 1)]N with R asthe dilution ratio and N as the number of subunits; since the PM domain induces a nearly irreversible membrane binding interaction, it wasassumed that one PM domain per oligomer is sufficient for inducing translocation).(B) The insertion of CaMKIIa into heterooligomers with mostly CaMKIIb is a stochastic process. The panel shows a plot of the relative plasmamembrane translocation of GFP–CaMKIIa at decreasing ratios of expressed PM–CaMKIIb. Each point is an average of at least ten experiments.The predicted curve for a stochastic insertion of PM–CaMKIIa into CaMKIIb oligomers overlaps with the fit of the data.(C) Relative cortical localization of GFP–CaMKIIa plotted as a function of increasing dilutions of CaMKIIb. Relative cortical localization isdefined as DPM/Iav, with DPM as the intensity difference between the PM and the cytosol and Iav as the average fluorescence intensity of aparticular cell. Each point is an average of at least ten experiments.(D) Measurement of the change in the diffusion coefficient as a function of an increasing dilution of CaMKIIb to GFP–CaMKIIa. The apparentdiffusion coefficient of GFP–CaMKIIa increased from 0.2 to 1 mm2/s as the ratio of CaMKIIb to CaMKIIa was lowered from 1:2 to 1:9. Theoutermost left and right data points show the diffusion coefficients of GFP–CaMKIIb and GFP–CaMKIIa, respectively.

cytoskeletal targeting of CaMKIIa by CaMKIIb, we mea- enzymes and signaling proteins are (1) increased effi-ciency and (2) increased specificity. By targeting CaMKIIsured the binding interactions of the heterooligomers

by measuring their diffusion coefficients. As shown in to dendritic spines, its local concentration is elevatedand the efficiency of substrate phosphorylation is in-Figure 1F, GFP–CaMKIIa has a 5-fold faster apparent

diffusion than GFP–CaMKIIb. Diffusion coefficients were creased. Important dendritic substrates of CaMKII in-clude AMPA receptors, NMDA receptors, SynGAP, andthen measured at increasing dilutions of GFP–CaMKIIa

to CaMKIIb. Similar to the results with the localization MAP2.Hence, by localizing CaMKII to dendritic spines, theto the cortical cytoskeleton, the diffusion coefficient of

CaMKIIa was reduced by 50% when the concentration kinase can more effectively exert its functions in synap-tic plasticity. The postsynaptically localized AMPA re-of CaMKIIb exceeded 15% of that of GFP–CaMKIIa

(Figure 7D). Together, these measurements show that ceptor has been shown to be an important substrate ofCaMKII, and phosphorylation by CaMKII increases theCaMKIIb is a potent targeting domain that can localize

a much larger number of CaMKIIa isoforms to the actin AMPA receptor conductivity 3-fold (McGlade-McCullohet al., 1993; Barria et al., 1997). This upregulation ofcytoskeleton.the AMPA receptor is thought to be one of the criticalfunctions of CaMKII in synaptic plasticity (Nicoll andDiscussionMalenka, 1995). The postsynaptically localized NMDAreceptor has also been shown to be a substrate forFunctional Importance of Docking CaMKII to F-Actin

in Dendritic Spines and Cell Cortex CaMKII (Omkumar et al., 1996), although the function ofthe NMDA receptor phosphorylation by CaMKII is notWhat is the functional advantage of a subcellular local-

ization of CaMKII? The main advantages of localizing well understood. Furthermore, the NMDA receptor has

Page 10: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

Neuron602

been shown to bind to a-actinin, an actin binding protein were coexpressed at different ratios (K. S., unpublishedpresent in PSDs (Wyszynski et al., 1997). A recent study data).also suggested that CaMKII has an important postsyn- The heterologous targeting mechanism is particularlyaptic role in inhibiting the activity of the PSD protein interesting since only 15% of CaMKIIb was needed toSynGAP, a Ras-GTPase activating protein that is likely bind 50% of CaMKII heterooligomers to the actin cy-important in regulating the dendritic MAP kinase path- toskeleton. Thus, cells can potentially control the local-way (Chen et al., 1998). ization of the predominant CaMKIIa isoforms by regulat-

The dendritic tubulin binding protein MAP2 is a major ing the local expression of CaMKIIb. The differentCaMKII substrate and is knownto form a bridge between relative expression levels of CaMKIIb during develop-actin and tubulin. Phosphorylation by CaMKII has been ment and in different neuronal cell types suggests thatshown to break its binding interaction with actin, which this targeting mechanism is physiologically importantis potentially important for a reorganization of the den- and may be responsible for a distinct subcellular local-dritic morphology (Vallano et al., 1986). An important ization of CaMKII in different cell types and periods ofrole of CaMKII in stabilizing dendritic branches has been development. A cell type–specific difference in CaMKIIsuggested in recent studies of neurons using expressed attachment is consistent with the earlier finding that acatalytically active CaMKII (Wu and Cline, 1998). much smaller fraction of CaMKII is found in the soluble

In addition to its postsynaptic functions, CaMKII may fraction of isolated cerebellum than in the soluble frac-also be relevant for regulation of the presynaptic actin tion of the forebrain (10% versus 50%) (Kelly and Vernon,binding protein synapsin 1. It is likely that the direct 1985; Kelly et al., 1987). This is paralleled by a ratio ofassociation of CaMKII with F-actin is important for the CaMKIIb toCaMKIIa of 4:1 and 1:3, respectively, in theseenhancement of synapsin 1 phosphorylation by CaMKII. two brain regions (Miller and Kennedy, 1985).Functionally, phosphorylation by CaMKII has been shown An intriguing piece in the puzzle is the observation thatto disable the synapsin–actin binding interaction and CaMKIIa can be locally translated in dendrites (Burgin etthereby enables synaptic vesicles to become available al., 1990). It suggests that a spatially distinct expressionfor secretion (Llinas et al., 1991; Benfenati et al., 1992). of CaMKIIa and CaMKIIb within a cell could be used to

Finally, biochemical evidence suggests that a signif- regulate the isoform ratio of the assembled heterooli-icant fraction of CaMKII is enriched in postsynaptic

gomer and thereby control the fraction of CaMKII activitydensities (PSDs). This structure has been shown to

associated with F-actin.contain a marked amount of CaMKIIa and a lesser

Since a variable fraction of expressed CaMKIIb isamount of actin and CaMKIIb in hippocampal neurons.

present as a spliced variant CaMKIIb9, we also testedInterestingly, CaMKIIb and actin are present in PSDs at

the localization of GFP-tagged CaMKIIb9. Interestingly,about the same molar concentration (Kelly and Vernon,

CaMKIIb9 showed a markedly lower F-actin localization1985), making it possible that CaMKII localization tothan CaMKIIb (K. S., unpublished data). Thus, the F-actindendritic spines is mediated by a direct CaMKIIb–actinlocalization of CaMKIIa/b heterooligomers can be regu-interaction. Furthermore, studies by McNeill and Colbranlated not only by the respective expression of CaMKIIa(1995) and Strack et al. (1997) have shown that CaMKII,and CaMKIIb but also by the alternative splicing ofonce autophosphorylated, can bind to at least two post-CaMKIIb.synaptic density proteins of 140 and 190 kDa. In light

Together, these results suggest that if CaMKIIa andof the data in our study, it is conceivable that CaMKIICaMKIIb are translated at the same time and in the sameoligomers are initially prelocalized to the PSD by theirlocation, they form random heterooligomers that arereversible actin binding interaction but then switch theirdocked to F-actin. When taking into account the lowerPSD binding partner or further translocate to PSDs fol-F-actin affinity of the alternatively spliced variant oflowing CaMKII autophosphorylation.CaMKIIb, a criticalnumber for docking CaMKII activity tothe actin cytoskeleton is the local expression of CaMKIIbRole of CaMKIIb As an F-Actin Targeting Moduleversus the sum of CaMKIIa plus CaMKIIb9.Our study shows that CaMKIIb functions as a targeting

module that localizes CaMKIIa/b heterooligomers todendritic spines and other F-actin rich regions. Where is

Heterologous Targeting Model for CaMKIIthe structural sequence motif for heterooligomerization?Could the cytoskeletal targeting of CaMKIIa by CaMKIIbEarlier studies suggested that the motif for oligomeriza-be anexample for a more general heterologous targetingtion of CaMKII is located at its C terminus (Yamauchi etmechanism (Figure 8)? This question is particularlyal., 1989). This domain is 78% identical between CaMKIIaintriguing since the family of CaMKII isoforms has re-and CaMKIIb. At least for CaMKIIa, a C-terminal, 135cently been expanded with the cloning of CaMKIIg andamino acid sequence of CaMKIIa was shown to be nec-CaMKIId as well as the identification of a large number ofessary and sufficient for oligomer formation (Shen andsplice variants including CaMKIIb9, CaMKIIaB,CaMKIIdA,Meyer, 1998). Deletions of more than 8 amino acids atand CaMKIIdB (Braun and Schulman, 1995). If a het-the C terminus and more than 21 amino acids at theerologous targeting mechanism would exist for all iso-N-terminal end of this short domain were sufficient toforms, CaMKIIa should also form heterooligomers withabolish oligomerization. Using the Pull-Out binding assayCaMKIIg and CaMKIId isoforms. Indeed, when we ex-as an in vivo approach, we now confirmed that minimalpressed PM–CaMKIIa together with GFP-tagged CaMKIIgC-terminal domains of CaMKIIa and CaMKIIb are indeedand CaMKIId isoforms, a direct binding interaction couldsufficient for oligomer formation. In these measure-be demonstrated using thePull-Out binding assay (K. S.,ments, constructs encoding GFP- and PM-tagged mini-

mal oligomerization domains of CaMKIIa and CaMKIIb unpublished data). Thus, the coexpression of CaMKIIa,

Page 11: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

CaMKII Targeting603

Figure 8. Heterologous Targeting Model forCaMKII

The model predicts that the localization ofthe predominant CaMKII isoform (CaMKIIa inmany neurons) is regulated by the relativeexpression level of CaMKIIb and possiblyother targeting CaMKII isoforms and splicevariants such as CaMKIIb9, CaMKIIdB andCaMKIIaKAP. The latter two isoforms maytarget CaMKIIa or other nonlocalized CaMKIIisoforms to the nucleus or sarcomere struc-tures in different cell types. CaMKIIa andother nonlocalized CaMKII isoforms wouldthen serve as “amplifiers” that enhance thelocal activity of CaMKII in cell-specific re-gions defined by the expression of the actual

targeting isoforms. Thus, the specificity and efficiency of substrate phosphorylation by CaMKII heterooligomers could be controlled by thelocal cotranslation of dedicated targeting isoforms.

In Vitro TranslationCaMKIIb, CaMKIIg, and CaMKIId can likely generateIn vitro translation with SP6 RNA polymerase was performed ac-functional heterooligomers that contain all possiblecording to the manufacturer’s protocol using a commercial kit (TNT-combinations of CaMKII isoforms.coupled reticulocyte lysate system, Promega). In vitro transcription

While all of these CaMKII isoforms have a conserved reactions were performed using mRNAs as templates. The relativeoligomerization domain, they contain a variable region molar concentration of the different translated proteins was calcu-

lated by calibration using [35S]Met incorporation and by countingthat is inserted between the oligomerization domain andthe number of methionines in the respective protein. Nonradioactivethe regulatory domain. At least for the CaMKIIaB andMet was used to obtain CaMKIIs and fusion constructs for the auto-CaMKIIdB isoforms, this insert includes a nuclear local-phosphorylation assay.ization sequence that targets these two CaMKII variants

to the nucleus (Srinivasan et al., 1994; Brocke et al.,Ca21-Dependent and -Independent Autophosphorylation

1995). Thus far, one example is known where the cata- of CaMKII and GFP Fusion Proteinslytic and regulatory domain has been deleted and only CaMKII autophosphorylation assays were performed as described

previously (Hanson et al., 1994). Briefly, CaMKII isoforms and fusiona core oligomerization domain (CaMKIIa) and a variableconstructs were autophosphorylated at 308C in 25 ml reactions con-region are left in the catalytically incompetent proteintaining 50 mM PIPES (pH 7.0), 10 mM MgCl2, 500 mM CaCl2, 600(Bayer et al., 1996; Sugai et al., 1996). This cardiac anM calmodulin, 50 mg/ml BSA, and 200 mM [g-32P]ATP (6000 cpm/KAP protein (CaMKIIaKAP) is localized to the sarcomerepmol). The Ca21-dependent autophosphorylation reaction was

structures and is likely to serve as a “pure” targeting started by adding in vitro translation product into the reaction mixmodule that lacks a catalytic domain of its own. Using and stopped by addition of EDTA (16.7 mM final concentration) 30

s later. To measure the extent of the Ca21-independent autophos-the different subcellular localization to the actin cy-phorylation, a 30 s reaction at high Ca21 was followed by a 120 stoskeleton, nucleus, and sarcomeres as examples, wesecondary incubation in the presence of added EGTA (3.3 mM finalpropose that many of the isoforms and splice variantsconcentration). In control experiments, 3.3 mM EGTA were includedhave a specific role in localizing CaMKII heterooligomersin the initial reaction mix. The reaction mix was then resolved by

to particular cellular sites. In this model, the selectivity SDS-PAGE and subjected to Phosphorimager analysis. The densi-and efficiency of CaMKII action can be controlled by tometry of bands were measured and corrected by the amount

of kinase, which was determined in a separate in vitro translationincreasing or decreasing the expression of a less abun-reaction with [35S]Met as described above.dant “targeting isoform” such as CaMKIIb. Taken to-

gether, the formation of large heterooligomers betweenIn Vitro Transcription and RNA ProcessingCaMKII isoforms and the presence of different CaMKIIIn vitro transcription and RNA processing were performed as de-“targeting isoforms” strongly suggests that localizationscribed previously (Yokoe and Meyer, 1996; Shen and Meyer, 1998).

of CaMKII through “heterologous targeting” is a general Briefly, in vitro transcription with SP6 RNA polymerase was per-mechanism to control the efficiency and specificity of formed according to the manufacturer’s protocol using a commer-

cial kit (mMESSAGE mMACHINE, Ambion). 10 mM EDTA was usedCaMKII function.to terminate the reaction. RNAwas purified by column chromatogra-phy (RNeasy column, Qiagen) followed by the addition of a poly(A)Experimental Procedurestail. Poly adenylation was carried out for 30 min at 378C in a 50 mlreaction mixture containing 40 mM Tris-HCl (pH 8.0), 10 mM MgCl2,Cloning of CaMKII Fusion Constructs2.5 mM MnCl2, 250 mM NaCl, 0.25 mg/ml RNA, 250 mM ATP, andThe cDNA for rat CaMKIIa, CaMKIIb, and CaMKIIb9 were generous5 U poly(A) polymerase (Life Technologies). The reaction was termi-gifts from Dr. Howard Schulman. The construction of the GFP–nated by addition of 20 mM EDTA. Unincorporated ATP and saltsCaMKIIa vector was described previously (Shen and Meyer, 1998).were removed by applying the mRNA to a RNeasy column. TheTo obtain the in vitro transcription vector for CaMKIIa without GFP,eluent was dried and mRNA was dissolved at 1 mg/ml in the electro-the CaMKIIa cDNA was amplified by PCR and cloned into the inporation buffer (5 mM KCl, 125 mM NaCl, 20 mM HEPES [pH 7.4],vitro transcription vector dSHiro3. DNA sequencing were performedand 10 mM glucose).to exclude PCR errors. GFP–CaMKIIb and CaMKIIb were also cloned

into the SHiro3 and dSHiro3 vectors using a similar PCR strategy.The construction of PM–GFP or Cys–GFP was described previously Cell Culturing and Electroporation

RBL 2H3 cells were maintained in Dulbecco’s Minimum Essential(Oancea et al., 1998). PM–CaMKIIa and PM–CaMKIIb were madeby replacing the GFP sequence with CaMKIIa and CaMKIIb coding Medium (DMEM) supplemented with 20% fetal bovine serum (Life

Technologies, Gaithersburg, MD) at 378C with 5% CO2. The cellssequence in the same SHiro3 vector.

Page 12: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

Neuron604

were plated at 5 3 104 cells/cm2 on glass cover slips and were were washed three times with PBS, and coverslips were mountedonto glass slides using buffered glycerol mounting medium.allowed to attach to the coverslip for a minimum of 3 hr. Hippocam-

pal neurons obtained from 2- to 4-day-old postnatal rats were cul-tured as described in Ryan and Smith (1995) and used 10 days to Fibroblast Transfection and Western Blotting Assay

NIH-3T3 cells were plated in 35 mm dishes at a density of 1.0 3 1053 weeks after plating. A self-built small volume electroporation de-vice for adherent cells was used for electroporation (Teruel and per dish and incubated overnight at 378C in a humid atmosphere

containing 5% CO2. Cells were transfected with 1.5–2.2 mg of pSRa–Meyer, 1997). For the transfection of neurons, modified versions ofthe device and buffer conditions were used (Teruel et al., submitted). CaMKIIb or pSRa–CaMKIIa or cotransfected with pSRa–CaMKIIb

and pSRa–CaMKIIa using lipofectin plus (GIBCO/BRL) accordingAfter transfection, the electroporation buffers were replaced withthe same culture medium. to manufacturer’s instructions. Cells were harvested 48 hr after

transfection and extracted with 10 mM Tris-HCl, 50 mM KCl, 0.1mM EGTA, 0.1% Triton, 2 mM PMSF, and 5 mg/ml aprotinin, leupep-Functional Labeling of Presynaptic Terminals with FM 4-64tin, and pepstatin at room temperature for 10 min and centrifugedFunctional presynaptic terminals were visualized with FM 4-64 (Mo-at 30,000 3 g for 30 min at 48C. Various fractions of the cell extractlecular Probes). FM 4-64 is similar in structure and properties towere resolved by electrophoresis on SDS-polyacrylamide gelsFM 1-43, but its longer wavelength emission spectra make it more(12%), transferred to nitrocellulose, and blotted with monoclonalsuitable for dual channel fluorescence microscopy in conjunctionanti-CaMKIIa or anti-CaMKIIb antibody (GIBCO/BRL). The mem-with GFP (Ziv and Smith, 1996). Cells were loaded with FM 4-64 bybranes were blotted using a secondary antibody conjugated toreplacing the saline in the imaging chamber with high potassiumhorseradish peroxidase and visualized by enhanced chemilumines-solution (100 mM KCL, 20 mM HEPES, 1.5 mM CaCl2, 30 mM NaCl,cence (ECL; Amersham).1.5 mM MgCl2 [pH 7.4], and 6 mM of FM 4-64) for 20 s and switch

back to a saline solution for 5–10 min. After collecting a digital imagePull-Out Protein–Protein Interaction Assayof the labeled field (such as that in Figure 2D, left), the cells werePolyadenylated mRNA was made as described above. In many ex-stimulated again by switching to the same high potassium solution.periments, mRNA species were mixed and used for electroporationThe spatial distribution of the active presynaptic terminals couldat a final concentration of typically 1 mg/ml total. The relative transla-then be determined from a difference image (e.g., Figure 2C, center).tion efficiency was determined by a separate in vitro translationreaction using the same mRNA as a template. Images of transfectedDiffusion AnalysisRBL or NIH-3T3 cells were taken on a Zeiss confocal microscopeThe diffusion coefficients were determined using an analysis by8–12 hr after electroporation. PMA (1 mM) was added and imageswhich a photobleached area is produced by a focused laser pulseof single cells were taken under the same configuration. Imagesand the fluorescence recovery is fit to sequential two-dimensionalwere taken before and after PMA addition and were analyzed usingGaussian distributions. Ratio images of the fluorescence distributionNIH image software. We defined a plasma membrane translocationafter the bleach pulse to the distribution before the pulse were usedfactor as DPM/Ipre (Figure 5E).for the analysis. This analysis follows at the same time the decrease

in the bleach amplitude and the increase in the bleach radius. As-Acknowledgmentssuming mass conservation, the resulting fluorescence distributions

were fit by:This study was supported by National Institute of Health grants GM-48113 and GM-51457. We wish to thank Dr. Howard Schulman forFn(x, y) 5 1 2 (Fo 3 ao

2/an2) 3 exp(2[(x 2 xo)2 1 (y 2 yo)2]/an

2),the CaMKIIa and CaMKIIb constructs, Dr. Margaret Titus for help

with x and y as the pixel coordinates, an as the radius of the bleach with the biochemical fractionation assay and the F-actin bindingdiameter in the nth image, and Fn(x, y) as the local relative fluores- measurements, Dr. Arturo Delozanne for insightful disscusion, andcence intensity. A least square fit routine was used to fit, at the Elena Oancea for providing the Cys–GFP construct. We would alsosame time, Gaussian profiles to all images in the time series. An like to acknowledge the support of Wen Chen and Dr. Thomasapproximate diffusion coefficient was then determined from a graph Stauffer.of the square of the radius, an

2, versus time. The diffusion coefficientcan be directly obtained from the slope of this graph (Dy/Dx 5 4 3 Received January 19, 1998; revised July 15, 1998.D; with D as the diffusion coefficient; Shen and Meyer, 1998).

ReferencesModel Calculations of a Stochastic Insertion of CaMKIIb

Barria, A., Muller, D., Derkach, V., Griffith, L.C., and Soderling, T.R.Subunits into Heterooligomers(1997). Regulatory phosphorylation of AMPA-type glutamate recep-The probability of having one or more subunits randomly insertingtors by CaM-KII during long-term potentiation. Science 276, 2042–into a heterooligomer is 1 2 (probability of having no subunit in-2045.serted). The probability of having none inserted is R/(R 1 1)N, with

R as the ratio of GFP–CaMKIIa to CaMKIIb and N as the number of Bayer, K.U., Lohler, J., and Harbers, K. (1996). An alternative, nonki-subunits. nase product of the brain-specifically expressed Ca21/calmodulin-

dependent kinase II alpha isoform gene in skeletal muscle. Mol.Cell. Biol. 16, 29–36.Immunofluorescence

NIH-3T3 cells, RBL cells, and hippocampal neurons were cultured Benfenati, F., Valtorta, F., Rubenstein, J.L., Gorelick, F.S.,on glass coverslips and transfected with mRNA encoding GFP– Greengard, P., and Czernik, A.J. (1992). Synaptic vesicle-associatedCaMKIIb fusion construct. Seven to eight hours after transfection, Ca21/calmodulin-dependent protein kinase II is a binding protein forthe cells were fixed for 10 min at 48C with 4% paraformaldehyde in synapsin I. Nature 359, 417–420.PBS (1.2 mM KH2PO4, 8.1 mM Na2HPO4, 138 mM NaCl, and 2.7 mM Bennett, M., Erondu, N., and Kennedy, M. (1983). Purification andKCl [pH 7.4]). NIH-3T3 cells and RBL cells were permeabilized for characterization of a calmodulin-dependent protein kinase that is5 min at 48C with 0.1% Triton in PBS. Hippocampal neurons were highly concentrated in brain. J. Biol. Chem. 258, 12735–12744.permeabilized for 10 min at 48C with 0.1% Triton. For F-actin stain-

Braun, A.P., and Schulman, H. (1995). The multifunctional calcium/ing, rhodamine phalloidin (Molecular Probes) was incubated withcalmodulin-dependent protein kinase: from form to function. Annu.the cells for 30 min at room temperature at a dilution of 1:300 inRev. Physiol. 57, 417–445.PBS. For the staining of postsynaptic densities, hippocampal neu-Brocke, L., Srinivasan, M., and Schulman, H. (1995). Developmentalrons were incubated with an monoclonal PSD-95 antibody (catalogand regional expression of multifunctional Ca21/calmodulin-depen-number 05–428, Upstate Biotechnology, Lake Placid, NY) overnightdent protein kinase isoforms in rat brain. J. Neurosci. 15, 6797–6808.at 48C at a dilution of 1:200 and then in secondary Cy3-labeled anti-

mouse antibody (catalog number 115–165–062, Jackson Immuno- Burgin, K.E., Waxham, M.N., Rickling, S., Westgate, S.A., Mobley,W.C., and Kelly, P.T. (1990). In situ hybridization histochemistry ofResearch, West Grove, PA) for 1 hr at room temperature. The cells

Page 13: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

CaMKII Targeting605

Ca21/calmodulin-dependent protein kinase in developing rat brain. Mayford, M., Wang, J., Kandel, E.R., and O’Dell, T.J. (1995). CaMKIIJ. Neurosci. 10, 1788–1798. regulates the frequency-response function of hippocampal syn-

apses for the production of both LTD and LTP. Cell 81, 891–904.Caceres, A., Payne, M., Binder, L., and Steward, O. (1983). Immuno-cytochemical localization of actin and microtubule-associated pro- Mayford, M., Bach, M.E., Huang, Y.Y., Wang, L., Hawkins, R.D., andtein MAP2 in dendritic spines. Proc. Natl. Acad. Sci. USA 80, 1738– Kandel, E.R. (1996). Control of memory formation through regulated1742. expression of a CaMKII transgene. Science 274, 1678–1683.

Chapman, P.F., Frenguelli, B.G., Smith, A., Chen, C.M., and Silva, McGlade-McCulloh, E., Yamamoto, H., Tan, S.E., Brickey, D.A., andA.J. (1995). The a-Ca21/calmodulin kinase II: a bidirectional modula- Soderling, T.R. (1993). Phosphorylation and regulation of glutamatetor of presynaptic plasticity. Neuron 14, 591–597. receptors by calcium/calmodulin-dependent protein kinase II. Na-

ture 362, 640–642.Chen, H.J., Rojas-Soto, M., Oguni, A., and Kennedy, M.B. (1998). Asynaptic Ras-GTPase activating protein (p135 SynGAP) inhibited by McNeill, R.B., and Colbran, R.J. (1995). Interaction of autophosphor-CaM kinase II. Neuron 20, 895–904. ylated Ca21/calmodulin-dependent protein kinase II with neuronal

cytoskeletal proteins. Characterization of binding to a 190-kDa post-Colbran, R.J., and Soderling, T.R. (1990). Calcium/calmodulin-inde-synaptic density protein. J. Biol. Chem. 270, 10043–10049.pendent autophosphorylation sites of calcium/calmodulin-depen-

dent protein kinase II. Studies on the effect of phosphorylation of Meyer, T., Hanson, P.I., Stryer, L., and Schulman, H. (1992). Calmod-threonine 305/306 and serine 314 on calmodulin binding using syn- ulin trapping by calcium-calmodulin-dependent protein kinase. Sci-thetic peptides. J. Biol. Chem. 265, 11213–11219. ence 256, 1199–1202.De Koninck, P., and Schulman, H. (1998). Sensitivity of CaM kinase Miller, S.G., and Kennedy, M.B. (1985). Distinct forebrain and cere-II to the frequency of Ca21 oscillations. Science 279, 191–192. bellar isozymes of type II Ca21/calmodulin-dependent protein kinase

associate differently with the postsynaptic density fraction. J. Biol.Egelhoff, T.T., Brown, S.S., and Spudich, J.A. (1991). Spatial andtemporal control of nonmuscle myosin localization: identification of Chem. 260, 9039–9046.a domain that is necessary for myosin filament disassembly in vivo. Miller, S.G., and Kennedy, M.B. (1986). Regulation of brain type IIJ. Cell Biol. 112, 677–688. Ca21/calmodulin-dependent protein kinase by autophosphorylation:Fisher, M., Kaech, S., Knutti, D., and Matus, A. (1998). Rapid actin- a Ca21-triggered molecular switch. Cell 44, 861–870.based plasticity in dendritic spines. Neuron 20, 847–854. Mukherji, S., and Soderling, T.R. (1994). Regulation of Ca21/calmod-Giese, K.P., Fedorov, N.B., Filipkowski, R.K., and Silva, A.J. (1998). ulin-dependent protein kinase II by inter- and intrasubunit-catalyzedAutophosphorylation of Thr286 of the calcium-calmodulin kinase II in autophosphorylations. J. Biol. Chem. 269, 13744–13747.LTP and learning. Science 279, 873–876. Nicoll, R.A., and Malenka, R.C. (1995). Contrasting properties of twoGlazewski, S., Chen, C.M., Silva, A., and Fox, K. (1996). Requirement forms of long-term potentiation in the hippocampus. Nature 377,for a-CaMKII in experience-dependent plasticity of the barrel cortex. 115–118.Science 272, 421–423. Nomura, T., Kumatoriya, K., Yoshimura, Y., and Yamauchi T. (1997).Gordon, J.A., Cioffi, D., Silva, A.J., and Stryker, M.P. (1996). Deficient Overexpression of alpha and beta isoforms of Ca21/calmodulin-plasticity in the primary visual cortex of a-calcium/calmodulin- dependent protein kinase II in neuroblastoma cells—H-7 promotesdependent protein kinase II mutant mice. Neuron 17, 491–499. neurite outgrowth. Brain Res. 766, 129–141.GuptaRoy, B., and Griffith, L.C. (1996). Functional heterogeneity of Oancea, E., Teruel, M.N., Quest, A.F.G., and Meyer, T. (1998). GFP-alternatively spliced isoforms of Drosophila Ca21/calmodulin- tagged cysteine-rich domains from protein kinase C as fluorescentdependent protein kinase II. J. Neurochem. 66, 1282–1288. indicators for diacylglycerol signaling in living cells. J. Cell Biol. 140,

485–498.Hanson, P.I., and Schulman, H. (1992). Inhibitory autophosphoryla-tion of multifunctional Ca21/calmodulin-dependent protein kinase Omkumar, R.V., Keily, M.J., Rosenstein, A.J., Min, K.T., and Ken-analyzed by site-directed mutagenesis. J. Biol. Chem. 267, 17216– nedy, M.B. (1996). Identification of a phosphorylation site for cal-17224. cium/calmodulin-dependent protein kinase II in the NR2B subunitHanson, P.I., Meyer, T., Stryer, L., and Schulman, H. (1994). Dual of N-methyl-D-aspartate receptor. J. Biol. Chem. 271, 31670–31678.role of calmodulin in autophosphorylation of multifunctional CaM Ryan, T.A., and Smith, S.J. (1995). Vesicle pool mobilization duringkinase may underlie decoding of calcium signals. Neuron 12, action potential firing at hippocampal synapses. Neuron 14,943–956. 983–989.Kanaseki, T., Ikeuchi, Y., Sugiura, H., and Yamauchi, T. (1991). Struc- Scholz, W.K., Baitinger, C., Schulman, H., and Kelly, P.T. (1988).tural features of Ca21/calmodulin-dependent protein kinase II re- Developmental changes in Ca21/calmodulin-dependent protein ki-vealed by electron microscopy. J. Cell Biol. 115, 1049–1060. nase II in cultures of hippocampal pyramidal neurons andKelly, P.T., and Vernon, P. (1985). Changes in the subcellular distri- astrocytes. J. Neurosci. 8, 1039–1051.bution of calmodulin-kinase II during brain development. Brain Res.

Schworer, C.M., Colbran, R.J., and Soderling, T.R. (1986). Reversible350, 211–224.

generation of a Ca21-independent form of Ca21(calmodulin)-depen-Kelly, P.T., Shields, S., Conway, K., Yip, R., and Burgin, K. (1987). dent protein kinase II by an autophosphorylation mechanism. J.Developmental changes in calmodulin-kinase II activity at brain syn- Biol. Chem. 261, 8581–8584.aptic junctions: alterations in holoenzyme composition. J. Neuro-

Shen, K., and Meyer, T. (1998). Identification of the minimal oligomer-chem. 49, 1927–1940.ization domain of Ca21/calmodulin-dependent protein kinase IIa. J.

Lai, Y., Nairn, A.C., and Greengard, P. (1986). Autophosphorylation Neurochem. 70, 96–104.reversibly regulates the Ca21/calmodulin-dependence of Ca21/cal-

Silva, A.J., Stevens, C.F., Tonegawa, S., and Wang, Y. (1992). Defi-modulin-dependent protein kinase II. Proc. Natl. Acad. Sci. USA 83,cient hippocampal long-term potentiation in alpha-calcium-calmod-4253–4257.ulin kinase II mutant mice [see comments]. Science 257, 201–206.

Landis, D., and Reese, T. (1983). Cytoplasmic organization in cere-Soderling, T.R. (1993). Calcium/calmodulin-dependent protein ki-bellar dendritic spines. J. Cell Biol. 97, 1169–1178.nase II: role in learning andmemory. Mol. Cell. Biochem. 128, 93–101.Llinas, R., Gruner, J.A., Sugimori, M., McGuinness, T.L., andSpector, I., Shochet, N.R., Blasberger, D., and Kashman, Y. (1989).Greengard, P. (1991). Regulation by synapsin I and Ca(21)-calmodu-Latrunculins novel marine macrolides that disrupt microfilament or-lin-dependent protein kinase II of the transmitter release in squidganization and affect cell growth. I. Comparison with cytochalasingiant synapse. J. Physiol. 436, 257–282.D. Cell Motil. Cytoskeleton 13, 127–144.Lou, L.L., Lloyd, S.J., and Schulman, H. (1986). Activation of theSrinivasan, M., Edman, C.F., and Schulman, H. (1994). Alternativemultifunctional Ca21/calmodulin-dependent protein kinase by auto-splicing introduces a nuclear localization signal that targets multi-phosphorylation: ATP modulates production of an autonomous en-

zyme. Proc. Natl. Acad. Sci. USA 83, 9497–9501. functional CaM kinase to the nucleus. J. Cell Biol. 126, 839–852.

Page 14: Neuron, Vol. 21, 593–606, September, 1998, Copyright 1998 ...teruel1/shen_neuron.pdf · threonine 286 (Shen and Meyer, 1998). An additional criterion for functionally intact GFP–CaMKII

Neuron606

Strack, S., Choi, S., Lovinger, D.M., andColbran, R.J. (1997). Translo-cation of autophosphorylated calcium/calmodulin-dependent pro-tein kinase II to the postsynaptic density. J. Biol. Chem. 272, 13467–13470.

Sugai, R., Takeuchi, M., Okuno, S., and Fujisawa, H. (1996). Molecu-lar cloning of a novel protein containing the association domain ofcalmodulin-dependent protein kinase II. J. Biochem. 120, 773–779.

Teruel, M.N., and Meyer, T. (1997). Electroporation induced forma-tion of individual calcium entry sites in cell body and processes ofadherent cells. Biophys. J. 73, 1785–1796.

Vallano, M.L., Goldenring, J.R., Lasher, R.S., and Delorenzo, R.J.(1986). Association of calcium/calmodulin-dependent kinase withcytoskeletal preparations: phosphorylation of tubulin, neurofila-ment, and microtubule-associated proteins. Ann. NY Acad. Sci. 466,357–374.

Wu, G.Y., and Cline, H.T. (1998). Stabilization of dendritic arborstructure in vivo by CaMKII. Science 279, 222–226.

Wyszynski, M.,Lin, J., Rao, A., Nigh, E., Beggs, A.H., Craig, A.M., andSheng, M. (1997). Competitive binding of a-actinin and calmodulin tothe NMDA receptor. Nature 385, 439–442.

Yamauchi, T., Ohsako, S., and Deguchi, T. (1989). Expression andcharacterization of calmodulin-dependent protein kinase II fromcloned cDNAs in Chinese hamster ovary cells. J. Biol. Chem. 264,19108–19116.

Yokoe, H., and Meyer, T. (1996). Spatial dynamics of GFP-taggedproteins investigated by local fluorescence enhancement. NatureBiotech. 14, 1252–1256.

Ziv, N.E., and Smith, S.J. (1996). Evidence for a role of dendriticfilopodia in synaptogenesis andspine formation. Neuron 17, 91–102.