<<

The Journal of Neuroscience, April 1995, 7~74): 32003214

Excitotoxic Activation of the NMDA Receptor Results in Inhibition of Calcium/CaImodulin Kinase II Activity in Cultured Hippocampal

Severn B. Churn, David Limbrick, Sompong Sombati, and Robert J. DeLorenzo Department of Neurology, Medical College of Virginia, Richmond, Virginia 23298

Neurotoxic effects of excitatory amino acids have been im- dependenton activation of the NMDA subtype of glutamatere- plicated in various neurological disorders, and have been ceptors (Choi et al., 1988; Koh and Choi, 1991) and has been utilized for excitotoxic models of delayed neuronal correlated with increasedintracellular calcium levels (Connor et death. The excitotoxic glutamate-induced, delayed - al., 1988; Glaum et al., 1990; Wadmanand Connor, 1992; Hart- al also results in inhibition of calciumlcalmodu- ley et al., 1993). In addition, removal of extracellular calcium lin-dependent kinase II (CaM kinase II). In this report, we from the culture media during glutamate exposure has a bene- characterized the glutamate-induced inhibition of CaM ki- ficial effect on neuron survival (Choi, 1987; Choi et al., 1988; nase II in relation to loss of intracellular calcium regulation Michaels and Rothman, 1990; Choi and Hartley, 1993). There- and delayed neuronal cell death. Glutamate (500 PM for 10 fore, alteration of neuronalcalcium physiology may be a major min), but not KCI (50 mM), exposure resulted in a significant mechanismby which glutamate exposure induces cell death. inhibition of CaM kinase II activity. The inhibition of CaM The multifunctional Ca2+/calmodulin-dependentprotein ki- kinase II activity was observed immediately following ex- naseII (CaM kinase II) is a major Ca*+ secondmessenger sys- citotoxic glutamate exposure and present at every time tem that regulatesmany Ca2+-dependent processes in neurons point measured. Glutamate-induced inhibition of kinase ac- (Jamesonet al., 1980; Caroni and Carafoli, 1983; Goldenring et tivity and delayed neuronal cell death was dependent upon al., 1984a; Yamamoto et al., 1985; Sakakibaraet al., 1986; Bas- both the activation of the NMDA sub- kys et al., 1990; Amador and Dani, 1991; McGlade-McGulloh type and the presence of extracellular calcium. The rela- et al., 1993). CaM kinase II phosphorylatesand regulatesthe tionship between inhibition of CaM kinase II activity and function of receptor-gatedion channels(McGlade-McGulloh et loss of intracellular calcium regulation was also examined. al., 1993; Machu et al., 1993), neuroskeleton elements (De- Experimental conditions which resulted in significant neu- Lorenzo et al., 1982), calcium-dependention channels(Sakaki- ronal cell death and inhibition of CaM kinase II activity also bara et al., 1986; Baskys et al., 1990) and is involved in neu- resulted in a long-term loss of intracellular calcium regu- rotransmission(DeLorenzo, 1981; DeLorenzo, 1983; Llinas et lation. Thus, inhibition of CaM kinase II activity occurred al., 1985). CaM kinaseII comprises1% of total forebrain under experimental conditions which resulted in loss of and up to 2% of total hippocampalprotein (Goldenring et al., neuronal viability and loss of neuronal calcium regulation. 1983; Kennedy et.al., 1983b; Ouimet et al., 1984; Erondu and Since the glutamate-induced inhibition of CaM kinase II ac- Kennedy, 198.5;Schulman, 1989). In addition, CaM kinaseII is tivity preceded neuronal cell death, the data support the predominantly expressedin neuronsrelative to glial cells (Eron- hypothesis that inhibition of CaM kinase II activity may play du and Kennedy, 1985; Schulman, 1989; Churn et al., 1992b). a significant role in excitotoxicity-dependent, delayed neu- The a-subunit is homologousto the major postsynaptic density ronal cell death. protein (PSD) which comprisesup to 50% of the total PSD [Key words: excitotoxicity, cell culture, calcium, phos- protein (Kennedy et al., 1983a; Goldenring et al., 1984b; Kelly phorylation, glutamate cations, pharmacology] et al., 1984). The high synaptic expressionof CaM kinase II suggeststhat this enzyme may be important for normal synaptic Neurotoxic effects of excitatory amino acids have been impli- function. Since CaM kinase II is a neuronally enrichedenzyme cated in various neurological disordersand have been utilized that regulatesmany important cellular processes,inhibition of for excitotoxic modelsof delayed neuronalcell death (Rothman, this important enzyme system would have important effects on 1986; Choi and Rothman, 1990; Choi, 1992). Glutamate-in- neuronal function. duced, delayed neuronal excitotoxicity has been shown to be Significant inhibition of CaM kinase II activity has been ob- served in many modelsof delayed neuronalcell death including Received Mar. 25, 1994; revised Oct. 18, 1994; accepted Nov. 10, 1994. whole animal models of (Taft et al., 1988; Churn et This work was supported by an NINDS Jacob Javits award (ROLNS23350) al., 1990b, 1992a; Zivin et al., 1991; Aronowski et al., 1992) and Epilepsy Program Proiect (POl-NS25630) to R.J.D., NIH Postdoctoral and glutamate excitotoxicity in neuronalcultures (Churn et al., Grant iHiO?537) 70 S.B.C, and the Sophie and Nathan Gumenick Neurosci- ence and Alzheimer Research Fund. We thank Adam M. Gray for his expert 1993). Transient forebrain ischemiaresults in greater than 50% technical assistance on the Syntide II assays. inhibition of CaM kinase II activity in hippocampus(Taft et al., Correspondence should be addressed to Severn B. Churn, Ph.D., Department 1988; Churn et al., 1990b)and cortex (Churn et al., 1990b). The of Neurology, Medical College of Virginia, Box 599 MCV Station, Richmond, VA 23298. decreasein CaM kinase II activity observed after ischemiais an Copyright 0 1995 Society for Neuroscience 0270-6474/95/153200-15$05.00/O early (within 10 set) and long-lastingphenomenon that precedes The Journal of Neuroscience, April 1995, 7~74) 3201 the development of delayed neuronal cell death (Taft et al., 1988; blue exclusion was used to determine neuronal viability. Surviving cells Churn et al., 1990b). This inhibition of CaM kinase II activity are expressed as percent viable cells remaining from original counts. has been implicated in contributing to delayed neuronal cell CaM kinase II assays death by several laboratories (Taft et al., 1988; Zivin et al., 1991; Under sterile conditions, culture medium was replaced with recording Aronowski et al., 1992; Churn et al., 1993). Therefore, under- solution as described above. Following experimental treatment, cultures standing the cellular mechanisms involved in regulating the in- were washed three times with recording solution and two times with hibition of CaM kinase II would provide insight into the culture medium. Neuronal cultures were allowed to recover in neuronal molecular mechanisms of excitotoxicity induced changes in neu- feed for specified times at 37”C, and isolated for quantitation of kinase activity. For quantitation of CaM kinase II activity, hippocampal neu- ronal transducing systems. rons were washed twice with recording solution at 37°C. The wash In this report, primary hippocampal neuronal cultures were solution was rapidly replaced with ice-cold homogenization buffer (100 utilized to characterize the effects of excitotoxic glutamate ex- mu piperazine-iV,N’-bis[2(-ethane sulfonic acid)] (PIPES) pH 6.9, 1 mM posure on delayed cell death, alteration of internal free calcium ethylenediamine-tetra-acetic acid (EDTA), 2 mu [ethylenebis(oxyethyl- regulation and inhibition of CaM kinase II activity. The data enenitrilo)] tetra-acetic acid (EGTA), 0.3 mu phenylmethylsulfonyl flu- oride), and the cells were immediately scraped from the culture dish indicate that excitotoxic glutamate exposure resulted in a sig- surface. The suspension was transferred into a glass homogenizer (Kon- nificant inhibition of CaM kinase II activity and that the ob- tes, Vineland, NJ), and disrupted as described previously (Churn et al., served inhibition of this enzyme is NMDA receptor mediated. 1992a) as modified in (Churn et al., 1993). Homogenates were nor- Inhibition of CaM kinase II activity occurred immediately fol- malized for protein and studied for endogenous calcium-dependent pro- lowing glutamate exposure and persisted until delayed neuronal tein phosphorylation. Standard phosphorylation reaction solutions con- tained 41 p,g of protein, 10 mM MgCl,, 7 FM y-‘*P ATP, 10 mM PIPES cell death occurred. In addition, inhibition of CaM kinase II pH 7.4, ?5 mM CaCl, and 2600 PM calmodulin. The concentration activity was dependent on extracellular calcium and could be of free calcium was estimated with a Ca2+/EGTA buffer system (Por- prevented by omission of calcium from the culture medium. The tzehl et al., 1964) as described in Burke and DeLorenzo (1981). The data support the hypothesis that inhibition of CaM kinase II standard free Ca2+ concentration for maximal activity was 50 FM. Final reaction volume was 100 pl. Standard reactions were performed in a activity may be involved in calcium-dependent, delayed neuron- shaking water bath at 30°C. Reactions were initiated by the addition of al cell death. calcium, continued for 1 min, and terminated by the- addition of 5% sodium dodecvl sulfate (SDS) STOP solution (Churn et al.. 1992a). Materials and Methods were iesolved by sodium dodecyl sulfate-polyacrylamide gel Hippocampal cell culture electrophoresis (SDS-PAGE) and protein bands visualized as described previously (Churn et al., 1992a). Stained gels were dried and exposed Primary hippocampal cultures were prepared by a modification of the to x-ray film (XRP-1, Kodak) for autoradiography. CaM kinase II sub- method of Banker and Cowan (1977) as described by Sombati et al. units were identified by running purified kinase fractions in parallel gel (1991). Briefly, hippocampal cells were prepared from 2 d postnatal rats lanes as described (Churn et al., 1990a, 1992a,b; 1993). The CaM ki- (Harlan) and grown on a confluent hippocampal astroglial feeder layer. nase II subunits were further identified by Western analysis (see below) Astrocytes were prepared from 2 d old pups by the methods of Abney of autophosphorylated protein using a monoclonal antibodies against et al., (1981). The glial cultures were maintained for 2 weeks in the 60 the B subunit (Churn et al., 1992b) or the o( subunit (Boehringer Mann- mm dishes (Costar) and fed twice weekly with MEM, 10% fetal bovine heim, Indianapolis) of CaM kinase II. The autoradiograph was then used serum, and 2 mM L-glutamine, 10 mu . After confluence (ap- as a template for excising radioactive phosphoproteins for quantitation proximately 4 d), the glial cultures were exposed to 5 pM cytosine in a liquid scintillation spectrometer (model LS 2800, Beckman), with arabinoside to inhibit further proliferation. The day prior to neuronal a counting efficiency of 80% (Churn et al., 1990a, 1992a). In some plating, the glial feed was replaced with N,-supplemented neuronal experiments,computer-assisted densitometry was utilized (Mocha,Jan- feed. The N, supplement contained 25 mu HEPES buffer (pH 7.4), 2 de1 Corp., San Rafael, CA), as previously described (Churn et al., mM glutamine, 5 kg/ml insulin, 100 bg/ml transferrin, 100 p,M putres- 1992a). Inhibition of CaM kinase II activity was analyzed by one-way tine, 30 PM sodium selenite, 20 pM , 1 mu sodium py- ANOVA (Graphpad, San Diego, CA). ruvate, 0.1% ovalbumin, 20 ug/ml T,, 40 kg/ml corticosterone. Hip- pocampal cells were plated at a density of 7.5 2 lo5 cells/60 mm culture dish onto a confluent glial bed. Cultures were maintained at 37°C under Phosphocelluloseassay of CaM kinase II phosphorylation of 5% CO,, 95% air. Culmres were fed three feedings/week (l/2 half media Syntide II change) with N,-supplemented, Earle’s Salt’s containing minimum es- Substrate phosphorylation was performed by a modification of the sential medium (MEM). The glutamine. MEM and HEPES buffer were method of Hashimoto and Soderling (1987). For kinase activity, stan- obtained from GIBCO’ (Gaithirsburg, MD). All other culture reagents dard reactions were performed as described above, except that 60 pM were obtained from Sigma Chemical Co. (St. Louis), except where spec- Syntide II (Sigma, St. Louis) was included in the mixture. This con- ified. centration of Syntide II was found to provide maximal phosphate in- corporation under standard conditions (see Fig. 8). The phosphorylation assays reaction was initiated by the addition of Ca*+, allowed to continue for Under sterile conditions, neuronal feed was replaced with three com- 1 min and stopped by the addition of 20 mM EDTA; 10 p,l of the assay plete changes of recording solution. Careful changes of the culture me- solution was -immediately blotted onto phosphocellulose filter paper, dium were performed to minimize trauma to cultured neurons and pre- P-81 (Whatman. Maidstone. England) as described oreviouslv (Hashi- vent drying of culture beds. All changes were performed under a sterile moto and Soderling, 1987). ‘Each reaction was quanhtated in uiplicate. hood by slow suction removal of old medium with simultaneous, slow P-81 filter paper was then washed three times in 50 mM phosphoric application of new medium. Recording solution contained (in mM) acid, rinsed with acetone, and allowed to air dry. Radioactive phosphate NaCl, 145; KCl, 2.5; CaCl,, 1 MgCl,, 1; D-glucose, 10; Na-HEPES, 10; was quantitated by scintillation counting as described above. glycine, 0.01; pH 7.3. The osmolarity was adiusted to 325 mOsm with &rose. Recording solution was replaced with recording solution con- Phosphatasestudies taining 500 IJ,M glutamate (or 100 FM NMDA) and 10 PM glycine for To determine the phosphatase activity towards CaM kinase II, removal 10 min. Glutamate toxicity was also contrasted with 50 mM KC1 ex- of phosphate from the (Y subunit of CaM kinase II was measured as posure (Glaum et al., 1990). Cultures were washed by three complete described previously (Churn et al., 1992a). Control CaM kinase II was changes of recording solution (no drug). Neuronal feed was replaced phosphorylated as described above, except that the phosphorylation re- and neurons allowed to recover for 3 d under the normal feeding sched- action was terminated by the addition of 20 mM EDTA ? exogenously ule. Viable cells were identified as phase-bright cells with intact pro- added phosphatases. Homogenates were incubated with a mixture of cesses (Churn et al., 1993). Nonviable neurons appeared phase dark and purified phosphatases 1 and 2A (PPC) or control buffer for 20 min at contained crenelated neuritic processes. In some experiments, trypan 30°C in buffer containing 10 mM PIPES (pH 7.4), 10 mM MgCl,, and 3202 Churn et al. - NMDA Excitotoxicity and CaM Kinase II Inhibition

Figure I. Photomicrographs showing nonaggregating hippocampal neuronal cultures before and following toxic glutamate exposure. A, Control hippocampal neurons exposed to culture medium for IO min and observed 24 hr after exposure. The neurons appear phase-bright with intact neuritic The Journal of Neuroscience, April 1995, 15(4) 3203

0.5 mu oL-dithiothreitol (DTT) (Churn et al., 1990a,b, 1992a). De- below). Estimation of the change in free [Ca*+], was determined by phosphorylation reactions were continued for 20 min and terminated by quantitating the ratio of Indo-l fluorescence changes of free Indo-l protein solubilization with an SDS solution. In some experiments, the versus calcium-bound Indo-l (see below). Culture medium was replaced reaction was terminated by the addition of EDTA/PPC mixture + 0.6 with several washings of recording solution as described for biochem- FM okadaic acid (OA), a selective PPC inhibitor. Phosphatase activity ical and survival studies. For experimental conditions, simultaneous re- was expressed as percent phosphate removed from maximally phos- moval and addition of recording medium + either 500 p,~ glutamate phorylated OLsubunit (Churn et al., 1992a). or 50 mM KC1 was performed. In some experiments, extracellular cal- To determine whether glutamate-induced inhibition was reversible cium was omitted or the selective NMDA antagonist, MK-801 (20 FM), with phosphatase treatment, homogenates from control and glutamate- was included as described in the figure captions. The specific [Caz+], treated cultures were pretreated with PPC as described above under imaging methodology is described below: conditions that were demonstrated to remove the majority of phosphate Cell loading with Indo-1. To load hippocampal neurons with Indo-1, groups due to autophosphorylation. In reactions where subsequent ki- neuronal feed was removed and replaced with Recording solution (145 nase activity was tested, the incubation mixture also contained 600 FM mM NaCl, 2.5 mM KCI, 10 mM HEPES, 10 mM glucose, 2 mM CaCl,, calmodulin. Phosphatase reactions were terminated by incubating sam- 1 mM MgCl,, pH 7.3, osmolarity adjusted to 325 with sucrose) con- ples with 0.6 pM okadaic acid (BioMol, Plymouth Meeting) for 1 min taining 1 LLM Indo-l AM. Loading of cells was oerformed at 37°C for at 30°C. Following PPC treatment, standard CaM kinase II reactions 45 minutes. Cells were then washeh three times with recording solution were performed in the presence of 0.6 PM okadaic acid to inhibit phos- and incubated for an additional 1.5 min to allow for the cleavage of the phatase activity as described previously (Churn et al., 1992a). Reactions Indo-l AM to the free acid form by cellular esterases. were terminated, proteins resolved by SDS-PAGE and incorporation of Microjluorometry. After neuronal cells were loaded with Indo-I, phosphate quantitated as described above. [Ca2+], was measured using the ratio program (image analysis mode) of a confocal ACAS 570 Interactive Laser Cytometer (Meridian Instru- Western analysis ments, Okemos, MI). Coverglass chambers were secured on the ACAS Proteins were resolved on SDS-PAGE under standard conditions and moving stage and analyzed using a 100X inverted oil immersion ob- blotted onto nitrocellulose. Nitrocellulose was immersed in TWEEN- jective lens (Olympus, Lake Success, NY). Step size was adjusted such phosphate-buffered saline solution (TPBS); 10 mM NaPO,, 0.9% NaCl, that multiple cells could be monitored simultaneously and analyzed in- pH 7.5) and 0.5% v/v TWEEN-20 (Sigma, St. Louis). The nitrocellulose dividually. Calcium transients were expressed as the average of multiple strip was incubated in TPBS containing a previously characterized an- cells. Confocal microscopy was utilized to ensure that calcium mea- ti-B CaM kinase II antibodv &G-3. monoclonal: Churn et al.. 1992a.b) surements were of hippocampal neurons. Using the manufacturer’s op- or’ anti-o antibody (Boehrmger Mannheim, Indianapolis) for’ 1 hr. Un: tics set for Indo- I, 100 mW of a 360 pm line of argon laser was used bound antibody was removed by three washings in TPBS. The blot was to excite the Indo-1. Photomultiplier strength was held constant in all incubated in a solution of biotinylated secondary antibody in TPBS. experiments, and a 1% neutral density filter was used to prevent Indo-l Excess secondary antibody was removed by three washings with PBS photobleaching. The ratio of emitted wavelengths at 405 pm (Indo-l- (no TWEEN-20). Labeled proteins were reacted with an alkaline phos- calcium complex) and 485 pm (free Indo-I) was monitored to indicate phatase staining kit (Vector, Burlingame). Development of bands was relative calcium concentration. Free [Ca*+], was estimated by compar- stopped by two changes of water. Developed strips were allowed to air ison of fluorescence ratio values to a calcium calibration curve. All dry and quantitation was performed by computer-assisted densitometry experiments were performed in recording solution at room temperature. (Mocha, Jandel Corn. San Rafael. CA). Eaual blottine of orotein was Calcium calibration curve. In order to convert fluorescent ratios to confirmed by protein staining utilizing ‘a sensitive gold staming proce- calcium concentration, an aqueous, in vitro calcium calibration curve dure (Churn et al., 1992a,b) of parallel strips of nitrocellulose (Bio-Rad, was performed according to the method recommended by Meridian In- Hercules). Strips of nitrocellulose were incubated in phosphate-buffered struments. Experimental parameters were consistent with those from saline as above supplemented with 0.3% TWEEN-20. Washed cell culture experiments, and the buffer used was designed to approxi- membranes were briefly rinsed in water and stained with AuroDye (Bio- mate intracellular conditions (100 mM KCl, 1 mM EGTA, 50 mM HE- Rad, Richmond) until band formation reached saturation. PES, pH 7.2). The curve was generated by measuring the change in fluorescence after adding known amounts of calcium to buffer contain- Immunocytochemistry ing 10 FM Indo-l pentapotassium salt (nonpermeant). Kd values of EGTA and Indo-l were stabilized by the high concentration of HEPES Hippocampal cells were plated onto a confluent glial feeder bed on 12 in the buffer and by reducing temperature to 21°C. EGTA correction of mm circular coverslips (Baxter, Columbia, MD) at a density 1 X lo4 the calcium calibration curve was performed using ACAS software (Me- cells/mm. The lower cell density was utilized to facilitate photography ridian Instruments, Okemos, MI) assuming Kd values of 0.151 pM for of single neurons within the population. Slides were subiected to ex- EGTA and 0.250 FM for Indo- 1, perimental conditions as described above and immediately fixed for 30 min with 5% paraformaldehyde diluted into 0.1% PBS. Fixed cells were Results washed, and permeablized for 1 hr with 0.1% saponin, 0.25% gelatin in PBS. Permeablized cells were exposed to lC3-3D6 (1:200 dilution Characterization of the cell culture population. Hippocampal into PBS/SAP/GEL) for I hr. Treated cells were exposed to biotinylated- cells were plated onto a confluent glial bed at a density of 7.5 secondary antibody (anti-mouse IgG, Vector) for 1 hour in PBS/SAP/ X lo5 cells/60 mm dish (Fig. 1). In mature cultures, viable neu- GEL. Cells were then treated with Texas red avidin (1:lOO dilution in rons could be distinguished from glial cells as large, phase-bright PBS/SAP/GEL. Vector) for 30 min. Cover slim were mounted onto I cells with long branching neuritic processes (Fig. 1A). In con- slides with DABCO mounting media and allowed to dry for 30 min. Slides were stored at 4°C in the dark until analyzed. Photomicroscopy trast, glial cells appeared flat and polygonal in morphology and was performed using a Nikon inverted microscope with original mag- did not develop an extensive network of axons or dendrites. nification of 40X. Approximately 30% of the neurons in the cultures were identi- fied morphologically as pyramidal neurons (Coulter et al., 1992). Measurement of intracellular calcium Exposure of cultures to 500 PM glutamate for 10 min resulted The ACAS confocal interactive laser cytometer was utilized to study in a reproducible, delayed neuronal cell death. Nonviable neu- intracellular calcium Ka2+1.) in hiuoocamoal neurons under identical rons exhibited fragmented processes (Fig. 1B) and did not ex- conditions utilized to‘inves&ate &M kin&e II activity and neuronal cell death. Neurons were preloaded with a fluorescent calcium binding clude the vital dye, trypan blue (data not shown). Cell mor- probe (Indo-1) and changes in cellular fluorescence levels were moni- phology was observed 24 hr following the exposure of cultures tored through whole cell imaging using ACAS confocal imaging (see to control, glutamate or glutamate plus MK-801 solutions. Ex- t processes. B, Hippocampal neurons 24 hr post 500 FM glutamate exposure (10 min). Hippocampal neurons appear necrotic with fragmented processes, (arrows denote neuronal soma). 3204 Churn et al. * NMDA Excitotoxicity and CaM Kinase II Inhibition

s 100 (Fig. 2). Depolarizing cultured neurons with 50 mM KCI for IO h min did not result in significant loss of viable neurons (4.2 + E 1.8% loss, n = 3). In addition, extending the KCI exposure to 8 60 min did not result in significant loss of viable neurons (4.5 ti 75 + 3% loss, n = 3). Previous studies from this laboratory have demonstrated that 50 mM KC1 treatment of the cultured pro- duced complete neuronal depolarization (Coulter et al., 1992). K These observations suggest that calcium entering through volt- g 50 age-gated calcium channels was not sufficient, alone, to cause v) = the delayed neuronal cell death. These observations also provid- ed a mechanism for inducing prolonged increases in intracellular 25 calcium which did not result in delayed neuronal cell death. The a, findings are consistent with the results from other studies (Choi, .-% 1988; Choi and Hartley, 1993; Churn et al., 1993) confirming that the effects of calcium on cell death depend upon the mech- '> 0 : 1 3 10 10 60 anism of calcium entry. Eflects of glutamate and depolarization on neuronal calcium Glutamate KCI levels. It is important to document the levels of intracellular free Exposure Time (min) calcium ([Ca?+],), under conditions utilized to quantitate neuron- al cell death and CaM kinase II inhibition. ACAS confocal anal- Figure 2. Effect of glutamate and KCl-depolarization on neuronal vi- ysis of Indo-l fluorescence was utilized to directly measure tran- ability 24 hr postexposure. Neurons were exposed to glutamate (500 sient increases in [Caz+], following pharmacologic stimulation pM, solid bars, 11 = 8) or KCI (50 mM, hatched bars, n = 3) for the with either 500 pM glutamate or 50 mM KCI solutions (Fig. 3). time indicated and allowed to recover for 24 hr as described in the Healthy neurons displayed basal [Caz+], levels between 100-150 Materials and Methods. Viable neurons expressed as the mean percent k SEM of original neurons/dish. **, p < 0.001 l-way ANOVA, Bon- FM. Cultures with neurons displaying basal [ Ca’ j, levels greater ferroni corrected. than 150 pM were not used for experimentation (Murphy and Miller, 1988). Excitotoxic (10 min) glutamate exposure resulted in an initial increase in [CaZ+], to greater than 2 FM (n = 20 citotoxic glutamate exposure (500 FM, for 10 min) resulted in neurons) followed by a sustained [Ca”], transient. In addition, 89.33 ? 0.73% loss of hippocampal neurons after 24 hr (Fig. the [Cazi], transient persisted after the glutamate solution was 2), confirming results reported previously (Choi et al., 1987; replaced with control solution. Coadministration of MK-801 Churn et al., 1993). Approximately 29.61 + 2.13% neuronal with the glutamate (Fig. 3C) significantly reduced the initial loss was observed after 1 hr of glutamate exposure. The acute [Ca2+], transient and all cells returned to normal [Ca?‘], levels toxic reaction to glutamate exposure confirms previously re- upon removal of glutamate (n = 8 neurons). To determine ported acute neuronal loss and is probably sodium or chloride whether the increase in [Ca2’], was due predominantly to influx dependent (Choi, 1987). Control cultures did not exhibit a sig- of extracellular Ca*‘, glutamate exposure was performed in the nificant loss of viable neurons (2.68 + 1.2% loss, n = 6). There- absence of extracellular Ca’+. Cultures were washed four times fore, any cell loss during the first 24 hr following glutamate with Ca*+-free recording solution and subjected to excitotoxic exposure was due predominately to glutamate treatment and not glutamate in the absence of Ca’+. Removal of extracellular cal- due to an artifact of handling the cultures. cium blocked both the initial rise in [Ca’+ 1, as well as the sus- The excitotoxic effect of glutamate on neuronal viability was tained [W*], transient (Fig. 30; y1 = 7 neurons). Thus, both the dependent upon both duration of glutamate exposure and glu- initial rise in [CaZ], and the glutamate-induced, sustained cal- tamate concentration (Fig. 2). Decreasing the time of neuronal cium transients were shown in our cultures and as described by exposure to glutamate resulted in decreased cell loss relative to others (Abele et al., 1990; Connor and Tseng, 1988) to be de- 10 min exposure after 24 hr (Fig. 2). Glutamate exposure for 1 pendent upon extracellular Ca2+ and stimulation of the NMDA min did not result in significant loss of viable neurons (6.55 ? subtype of glutamate-gated channels. Exposure of neurons to 50 3.23% loss, y1 = 8). Glutamate exposure for 3 min resulted in a mM KCI for IO min (Fig. 3B) resulted in an initial increase in 47.3 I ? 2.9% loss compared to control treatment. The percent LCaZ+],, to approximately 2.4 pM with a smaller [Ca* ’ 1, transient cell death for the 3 min exposure was significantly different from that rapidly returned to baseline levels following removal of KCI both control and 10 min exposure times (p < 0.001, one-way solution (n = 9 neurons). Thus, stimulation of neurons with 50 ANOVA, Bonferroni corrected). Reducing glutamate concentra- mM KCI resulted in an initial peak of [Caz’], which was similar tion to 50 FM modulated the level of neuronal cell death. Ex- to glutamate exposure, but did not produce a sustained, elevated posure of neurons to 50 HAM glutamate for 3 min did not result [Ca?+], transient as was observed with 500 FM glutamate. in significant neuronal cell death (14.09 ? 1.4% loss, II = 8). IZects of glutamate exposure on CaM kinase II activity. Ho- However, 10 min of exposure to 50 pM glutamate resulted in a mogenates from control hippocampal neuronal cultures were re- significant loss of viable neurons (79.4 -t 2% loss, II = 9). Thus, acted under standard conditions (see Materials and Methods) the glutamate-induced, delayed neuronal cell death was depen- which were maximal for CaM kinase II activity and resolved by dent on both glutamate concentration and exposure duration. SDS-PAGE (Fig. 4). Homogenates from control cultures dem- To determine whether depolarization and entry of calcium onstrated calcium-stimulated phosphate incorporation into spe- through voltage-dependent channels could account for the glu- cific protein bands in comparison to magnesium-dependent (bas- tamate-induced neuronal cell death, cultures were treated with al) phosphorylation. The calcium-dependent phosphorylation SO mM KC1 under analogous conditions to glutamate exposure pattern observed in hippocampal cultures was similar to that The Journal of Neuroscience, April 1995, 75(4) 32aO5

Glutamate KC!

0 5 10 15 20 Time (min) Time (min)

C D GLU + MK-801 GLU + 0 Ca2+ 3 s 32.- r 3 1 - 0 L 0 0 5 10 15 20 0 5 10 15 20 Time (min) Time (min) Figure 3. Effect of glutamate and KCl-depolarization on [Ca2+li in hippocampal neurons in culture. Neurons cultured for 12 d were used for all experiments. Neurons were preloaded with Indo-l under standard conditions (see Materials and Methods) and transferred to the confocal fluorescence microscope. Solutions were rapidly added and removed with a flow through system that did not affect the cells. Cultured neurons were constantly in view during all experimental manipulations. The neurons were exposed for 10 min to, in A, 500 PM glutamate, n = 20; B, 50 mM KCl, n = 9; C, 500 p,~ glutamate + MK-801, n = 8; and D, 500 PM glutamate in the presence of 0 [Caz+]O. n = 7 neurons. After removal of the treatment conditions, neurons were maintained in recording solution for continued observation. The bar in each figure denotes the time of treatment application. ACAS confocal imaging of Indo-l fluorescence (see Materials and Methods) was utilized to estimate free [CaZ+],. Molar [CaZ+li levels were determined by comparing fluorescence intensity with a calibration curve (Meridian Instruments, Okemos). Figures show the average [Ca*+], values obtained from multiple recordings (n values given above). observed in preparationsfrom whole , hippocampalslices, the absence of neurons when incubated under standard condi- and from mixed cortical neuronalcultures (Churn et al., 1990a, tions utilized to measure kinase activity in neuronal cultures 1992a, 1993).Although CaM kinaseII activity hasbeen reported (data not shown).Thus, CaM kinase II activity observed in neu- in glial cells (Babcock-Atkinson et al., 1989), no significant ronal culture was primarily expressed in neuronal cells and was CaM kinase II activity was observed in glial cells cultured in not from glial cell contamination. Comigration with purified 3206 Churn et al. * NMDA Excitotoxicity and CaM Kinase II Inhibition

Control Glutamate Ca2+ l-1 (+I l-1 (+I **

$ 25 l-n 60 kDa . .-s I 50 kDa . 2 0 Control 1Glut KCI Treatment

Figure 5. Effect of glutamate (500 FM) or KC1 (50 mM) on CaM kinase II activity in hippocampal homogenates. Hippocampal neurons in culture were exposed to glutamate (II = 1 I) or KCI (n = 12) for 10 min under standard conditions and allowed to recover for I hr as de- scribed in Materials and Methods. The cultures were then harvested and endogenous kinase activity was quantitated as described in Materials and Methods. Endogenous kinase activity was calculated as percent of kinase activity in parallel sham-treated cultures. Data are presented as Figure 4. Effect of glutamate on endogenous CaM kinase II activity mean 2 SEM. **, p < 0.001, one-way ANOVA, Bonferroni corrected. in hippocampal neurons. The autoradiograph from a representative so- dium dodecyl sulfate-polyacrylamide gel electrophoresis of hippocam- pal culture homogenates reacted under standard conditions for CaM [Ca?‘], resulted in significant inhibition of CaM kinase II activ- kinase II activity (see Materials and Methods). Endogenous phosphor- ity. Conversely, depolarizing conditions that did not result in ylation was measured in the absence (-) and presence (+) of calcium significant neuronal death or prolonged elevations in [CaZ+],, and calmodulin as described in Materials and Methods. AW~WS denote the (Y(50 kDA) and p (60 kDA) subunits of CaM kinase II. CaM kinase also did not result in significant inhibition of CaM kinase II II activity was routinely measured by quantitation of a subunit auto- activity. phosphorylation (see Materials and Methods). To determinethe time courseof excitotoxic glutamate recep- tor activated inhibition of CaM kinase II activity, hippocampal neuronswere assayedfor kinase activity after different time in- CaM kinase II on high resolution gel electrophoresishas been tervals following a standard IO min glutamate exposure. After utilized to identify the 50 kDa (a) and 60 kDa (p) subunitsof excitotoxic exposure,significant inhibition of CaM kinaseII ac- CaM kinase II in forebrain homogenatesand neuronal cultures tivity was observed at all time points measured(Fig. 6). CaM (Churn et al., 1990a, 1993). Comigration with purified rat CaM kinase II inhibition was observed immediately following exci- kinaseII was utilized to identify the 50 kDa and 60 kDa subunits totoxic exposure to glutamate (46.1 -t 6.9% inhibition, II = 6 as the alpha and beta subunitsof CaM kinase II in hippocampal cultures) and remained diminished at all points measured.The cell culture (Fig. 4, arrows). The peptide bandsin the cellular data suggestthat excitotoxicity-induced inhibition of CaM ki- homogenatecomigrating with purified CaM kinase II were fur- naseII activity occurred, it remained until significant neuronal ther demonstratedto be the 01and p subunitsof the kinase by death was observed.Thus, glutamate-inducedexcitotoxicity was immunoreactivity with specific antibodiesdirected againstthese observed immediately following a 10 min glutamate exposure subunitsby Westernanalysis (see Materials and Methods). and at every time point measured,up to 1 hr. In addition, the Excitotoxic glutamateexposure that producedsignificant neu- observed inhibition of CaM kinase II activity precededthe glu- ronal death after 24 hr (Fig. 2) and elevated [CaZ+],(Fig. 3) in tamate-induced,calcium-dependent neuronal cell death. hippocampalcells resultedin a 41 ? 1.9% inhibition of CaM To further determine whether the observed inhibition of ki- kinase II activity when assayed 1 hr after glutamate exposure nase activity was due to autophosphorylationwith cold phos- (Fig. 5). The observed glutamate-inducedinhibition was signif- phate groups, CaM kinase IlLdependent phosphorylation of ex- icant when compared to control cultures (y < 0.01, one-way ogenously-addedsynthetic peptide (Syntide II) was performed ANOVA, Bonferroni corrected). Short-term exposure (l-3 min) (Fig. 7), utilizing establishedprocedures (Hashimoto and Sod- of hippocampalcells to glutamate did not result in significant erling, 1987). Under standardizedconditions, Syntide II phos- inhibition of CaM kinase II activity (data not shown). In addi- phorylation was inhibited in fractions obtainedfrom glutamate- tion, exposure of cultures to 50 mM KC1 for 10 min did not treated cultures (68.1 2 1% of control, 11 = 4). In addition, the result in inhibition of CaM kinaseII activity (97% of control, n inhibition observed in the Syntide II phosphorylationassays was = 12 cultures; Fig. 5). Thus, glutamate exposuresthat resulted equivalent to the inhibition observed in the back phosphoryla- in significant delayed neurotoxicity and prolongedelevations in tion assays(68.8 + 2.3, II = 4). Therefore, the glutamate-in- The Journal of Neuroscience, April 1995, 75(4) 3207

” 2 IOO- - - E 8 E 80- 8 **** k 60- V

.--2 40-

z: 20-

.-c v 0-L-L - - Sham Glutamate1 Sham GlutamateI E @--J-J- Subunit Syntide II 9 Contro’0.0 10 30 60 Phosphorylation Measurement 030) Figure 7. Glutamate-induced inhibition of CaM kinase II activity is Postexposure Time (min) equivalent when measured by autophosphorylation of the c1 subunit and phosphorylation of exogenously added substrate. Glutamate-induced in- F&w~ 6. Time course of glutamate-induced inhibition of CaM kinase hibition of CaM kinase II activity was measured by standard autophos- II activity. Cultures were exposed to control or glutamate solutions fat phorylation methods (n = 4) and P-81 assay of syntide II phosphory- IO min and harvested at the time point indicated. The excitotoxic glu- lation in the same samples (see Materials and Methods). Glutamate tamate exposure which induced delayed neuronal cell death also in- induced inhibition of kinase activity was equivalent when measured by duced an immediate inhibition of CaM kinase II activity which persisted either technique. The data represent the mean ? SEM of control kinase for all time points measured. Data represent mean +- SEM of control activity. **, p < 0.001, one-way ANOVA, Bonferroni corrected. kinase activity (M = 6, each group).

equilibrium of the phosphorylated and nonphosphorylated states duced inhibition of CaM kinase II activity observed in the back phosphorylation assays was due to inhibition of enzyme activity which may complicate the interpretation of in vitro phosphory- lation assays (Patton et al., 1993). Although the exogenous sub- and not due to prelabeling with cold phosphate groups of the strate studies performed above strongly rule out autophosphor- kinase subunits. To further characterize Syntide II phosphorylation, saturation ylation as the cause of glutamate-induced inhibition of kinase experiments were conducted. The glutamate induced inhibition autophosphorylation, it may still be possible that potential phos- of CaM kinase 11 activity could not be overcome by either in- phorylation sites were already phosphorylated in glutamate- creasing Syntide II concentration or time of reaction (Fig. 8). treated samples and thus not available for phosphorylation, in vitro. To determine whether de now autophosphorylation of Syntide II phosphorylation displayed an apparent k, of I I .4 PM CaM kinase II had occurred, homogenates from control and glu- in control homogenates and IO.2 pM in glutamate treated cul- tures. However, maximal Syntide II phosphorylation in gluta- tamate-treated cultures were treated with phosphatase I and 2A mate-treated cultures was decreased 36.9% when compared to (PPC) under conditions that were shown to effectively remove control phosphorylation (Fig. 8). In addition, the glutamate-in- endogenous phosphates from CaM kinase II (Shields et al., duced inhibition of Syntide II phosphorylation could not be 1985; Churn et al., 1992a). overcome by increasing reaction time (Fig. 8). The k,,, was not To determine whether phosphatases 1 and 2A, could remove significantly different in glutamate-treated cultures when com- autophosphorylation-dependent phosphate groups, autophos- pared to control (control k,,2 = 3 I .2 set vs glutamate-treated k,,2 phorylation reactions were performed and terminated by the ad- = 38 set). However, the maximal Syntide II phosphorylation in dition of PPC mixture. Removal of radioactive phosphate groups glutamate-treated cultures did not approach control phosphory- was quantitated over time by removing aliquots and resolving lation levels at any time point measured. Maximal Syntide II the proteins by SDS-PAGE (Fig. 9). Partial dephosphorylation phosphorylation was 45.6% of control phosphorylation using of endogenous kinase subunits was observed in buffer alone. saturating levels of substrate (Fig. 8). Thus, glutamate-induced The observed dephosphorylation was most likely due to endog- inhibition of CaM kinase II activity was observed when utilizing enous phosphatases in the homogenate fractions. The dephos- autophosphorylation as well as exogenously added Syntide II as phorylation of CaM kinase II was more rapid and occurred to a a substrate. In addition, the glutamate-induced inhibition of CaM significantly greater extent in the presence of exogenously added kinase II activity could not be overcome by increasing either PPC (Fig. 9) removing greater than 70% of labeled phosphate Syntide II concentration or time of reaction. groups added by autophosphorylation. Incubation of homoge- Glutumate-inducedinhibition of CUM kinase II was not due nates with both PPC and the selective phosphatase inhibitor oka- to autophosphor?ilation.Autophosphorylation has been shown daic acid (600 pM) resulted in almost no removal of radioactive to modulate CaM kinase II activity (Lai et al., 1986; Hanson phosphate groups from the CaM kinase II subunits. Thus, oka- and Schulman, 1992). In addition, CaM kinase II exists in an daic acid sensitive phosphatases were able to remove a signifi- 3208 Churn et al. * NMDA Excitotoxicity and CaM Kinase II Inhibition

A ‘u 100 $ o 80 .-E 1400 E 2 1200 $? 60

3 1000 2 jj 40 a 800 r 20 9 800 5 0 A. B 400 E O 5 200 Buffer PPC PPC

z 0 FI I I I I I I 0 20 40 60 80 100 120 140 O’A Figure 9. Activity of PPC in removing CaM kinase II phosphate Syntide II (PM) groups due to autophosphorylation. Homogenates from control hippo- campal cultures were phosphorylated as described in Materials and Methods. Autophosphorylation reactions were terminated by the addi- tion of chelator buffer + PPC or PPC + OA. PPC reactions were continued for 20 min and terminated by solubilization of reactant pro- B teins. Samples were resolved by SDS-PAGE and phosphate removal from the 01 (50 kDa) subunit quantitated by liquid scintillation counting (see Materials and Methods). Data are expressed as the mean percent t- SEM phosphatase removed from CaM kinase II compared to zero time point (n = 4, each group). **, p < 0.001 different from control .-G 2500 buffer solution, one-way ANOVA, Bonferroni corrected. 2 b 2000 phatasetreatment. Kinase from glutamate-treatedcultures dis- 2 1500 played 60.3% of control activity prior to phosphatasetreatment v and displayed61.8% activity following treatment.Thus, the glu- 2 1000 tamate-inducedinhibition of CaM kinase II was not due to in- creasedautophosphorylation of the enzyme in situ. Since no 5 500 kinase activity could be recovered by the PPC treatment, it is unlikely that autophosphorylation,alone, could account for the z 0 glutamate-inducedinhibition of CaM kinaseII activity. Together I with the substratephosphorylation data, these results indicated 0 2 4 6 8 10 12 that excitotoxic inhibition of CaM kinase II was not due to an autophosphorylation-inducedinhibition of the enzyme. Howev- Time (min) er, the data doesnot exclude the possibility that phosphorylation Figure 8. Syntide II phosphorylation in homogenates obtained from at a site not recognized by the phosphatasesused in the present control (squares)and glutamate-treated (triangles)cultures. A, Syntide study may accountfor someof the observed inhibition of kinase II saturation curves were generated in homogenates from control and activity. glutamate-treated cultures. Significant inhibition of kinase activity was Pharmacology of glutamate-inducedneurotoxicity. To deter- observed in glutamate-treated cultures (V,,, = 1437.4 fmol/pg/min con- trol vs 909.1 fmol/p,g/min, glutamate-treated) without alteration of k, mine which glutamatereceptor subtype was responsiblefor the (11.4 PM control vs 10.2 pM, glutamate-treated) when compared to excitotoxicity-induced inhibition of CaM kinaseII activity, spe- control cultures. B, Syntide II phosphorylation time courses for homog- cific glutamatereceptor agonistsand antagonistswere employed enates obtained from control and glutamate-treated cultures. Increasing (Fig. 10). Coincubation of neuronswith glutamate + MK-801, reaction time did not overcome glutamate-induced inhibition of CaM kinase II activity when measured by Syntide II phosphorylation. Glu- a noncompetitiveNMDA ,resulted in almost tamate treatment did not result in alteration of k,,, (31.2 set, control vs complete protection from the glutamate-mediatedinhibition of 38 set, glutamate treated). However, increasing reaction time did not CaM kinase II (5.35 + 4.0% inhibition, R = 4). Alternatively, overcome the glutamate-induced inhibition (V,,,,, 298 1 fmollpg, control incubation with 100 FM NMDA resulted in significant (60 + vs 1621 fmol/p,g, glutamate treated). Data shown are representative of 5.6%) inhibition of CaM kinase II activity. Furthermore, coin- multiple experiments. The data represent the mean 2 SEM of triplicate quantitations. cubation of neuronswith glutamate+ CNQX (100 FM), a potent AMPA receptor antagonist,did not result in significant protec- tion from glutamate-inducedinhibition of CaM kinaseII activity cant level of phosphategroups due to autophosphorylation of (52.7 ? 4% inhibition, n = 4). Incubation with quisqualate, CaM kinase II. resultedin 33.72 -t 10.17% inhibition of CaM kinaseII activity Unlike autophosphorylation,the excitotoxicity-induced inhi- (n = 4). However, the observed inhibition could be blocked with bition of CaM kinase II activity was not reversible with phos- coincubation of quisqualate+ MK-801 (9.17 2 2.39% inhibi- The Journal of Neuroscience, April 1995, 15(4) 3209

” 0 - g 100-l U I

8 25

.-i! - 2 0 s mg! Control 0.0 0.3 1.0 2.0 9 2.0 + .-s Ca2+ Concentration (mM) zi 5 .-u) b Figure 11. Effect of calcium on glutamate-induced inhibition of CaM 6 kinase II activity. Hippocampal neurons were exposed to standard ex- citotoxic glutamate (500 PM, 10 min) in the presence of varying extra- Figure 10. Pharmacology of glutamate-induced inhibition of CaM ki- cellular calcium concentrations. Removal of calcium resulted in protec- nase II activity in hippocampal cultures. Hippocampal cultures were tion from glutamate-induced inhibition. Data are expressed as mean * exposed to agents specified under standard excitotoxic conditions. Cul- SEM (n = 6). p < 0.001, one-way ANOVA, Bonferroni corrected. tures were allowed to recover for 1 hr and harvested for CaM kinase II quantitation. Endogenous kinase activity is expressed as the mean percent t SEM of sham-treated sister cultures; n = 4, each group. **, p < 0.01, one-way ANOVA, Bonferroni corrected. the presence of glutamate, resulted in similar levels of CaM kinase II inhibition (36.1 5 2.9; 31.2 & 2.3; 44.2 t- 1.2% inhibition, respectively). The results were similar to previously tion, II = 4). Thus, the observed quisqualate induced inhibition published observations of cation dependence on END (Coulter of CaM kinase II activity probably was due to endogenous glu- et al., 1992). Thus, the cation dependence for inhibition of CaM tamate release and secondary stimulation of NMDA-dependent kinase II activity parallels that observed for neurotoxicity (Choi receptors. The data support the hypothesis that excitotoxic ac- et al., 1988; Michaels, Rothman, 1990) and the induction of tivation of the NMDA glutamate-receptor subtype can account END (Sombati et al., 1991; Coulter et al., 1992) in primary for the excitotoxicity induced inhibition of CaM kinase II ob- neuronal cultures. served in this model. Immunocytochemistryof CaM kinase II in neuronal culture. Calcium dependenceof excitotoxicity-induced inhibition oj Our laboratory has developed a monoclonal IgG, antibody CUM kinase II activity. Previous reports have demonstrated the against the beta subunit of CaM kinase II which does not rec- dependence of calcium on delayed neuronal toxicity (Choi et al., ognize the ischemically modified enzyme in situ (Churn et al., 1988; Michaels and Rothman, 1990) and the development of an 1992a,b). This antibody was utilized to further characterize the extended neuronal depolarization (END, Coulter et al., 1992; inhibition of CaM kinase II activity in hippocampal cultures. Sombati et al., 1991). Therefore, we investigated whether a cal- Immunocytochemical analysis was performed on neurons ex- cium-dependent mechanism was responsible for the NMDA-in- posed for 10 min to either control or glutamate solution (Fig. duced inhibition of CaM kinase II activity. To assess the effect 12). Control cultures exhibited significant immunofluorescence of removal of calcium from the recording solution, cultures were which was specific to neuronal cells. The specific immunofluo- washed with physiological saline without calcium. The cultures rescence demonstrated the high level of neuronal CaM kinase II were then exposed to glutamate as described in the methods expression relative to glial cells in culture. In glutamate-treated except that calcium was omitted in the glutamate-containing re- cultures, many neurons did not show significant immunofluores- cording solution. In addition, calcium was omitted from the ini- cence above background levels. In the remaining neurons that tial washing steps after glutamate exposure. Removal of calcium did show immunofluorescence, the level was significantly lower from the recording solution resulted in almost complete protec- than control neurons (Fig. 12). In addition, the pattern of im- tion from glutamate-induced inhibition of CaM kinase II activity munofluorescence was grainy and not homogenous compared to (Fig. 1 1). A calcium concentration curve was tested to determine control cultured cells. Pyramidal neurons identified by mor- the effect of various extracellular calcium on inhibition of CaM phology demonstrated a more significant decrease in CaM ki- kinase II activity. Removal of extracellular Ca*+ during the glu- nase II immunoreactivity than other neuronal cell types. Thus, tamate exposure, prevented the glutamate-induced loss of CaM excitotoxic glutamate exposure resulted in a loss of immunoflu- kinase II activity (0.2 + 1.1% inhibition, n = 6). However, orescence that was observed immediately following excitotoxic extracellular CaZ+ concentrations of 0.3, 1, and 2 mM CaCl, in exposure of neuronal cultures to glutamate, confirming that glu- 3210 Churn et al. * NMDA Excitotoxicity and CaM Kinase II Inhibition

Figure 12. CaM kinase II immunocytochemistry in hippocampal neurons in cultures exposed to control (A, C) or glutamate (500 pM; B, D) for 10 min and immediately fixed with paraformaldehyde. Neurons were immunoreacted with a monoclonal IgG, (lC3-3D6) directed against the beta The Journalof Neuroscience,April 1995,15(4) 3211 tamate rapidly induced a significant inhibition of CaM kinase II the hippocampus(Churn et al., 1992b). Thus, altered immuno- activity following excitotoxic exposure. In addition, the de- reactivity was observed in neuronal populations that displayed creased immunoreactivity observed in situ confirmed that mea- delayed neuronal cell death, but not in populations that were surementof neuronal kinase, relative to glial kinase, is affected insensitive to the effects of transient ischemia.The same anti- by excitotoxic glutamate exposure. body was utilized to establishthe effect of glutamatetoxicity on CaM kinase II immunoreactivity in hippocampalcultures. Ex- Discussion citotoxic glutamate exposure resulted in an immediate loss of Primary hippocampalneuronal cultures were utilized to measure immunoreactivity of CaM kinase II with the monoclonal anti- directly the effect of glutamate exposure on neuronal viability, body. The decreasedimmunoreactivity paralleledthe decreased modulation of [Ca”],, and inhibition of CaM kinase II activity enzymatic activity observed in this model and was observed within the same preparation. Excitotoxic glutamate exposures prior to neuronalcell death. In the gerbil model of ischemia,the that resulted in delayed neuronal cell death and prolonged hippocampalpyramidal neurons which were sensitive to isch- [Ca*+], transientsalso resulted in significant inhibition of CaM emia are the samepopulations that lose immunoreactivity with kinase II activity. The glutamate-inducedinhibition of CaM ki- kinase antibody (Churn et al., 1992b). In the hippocampalcul- naseII activity was observed immediately following excitotoxic ture model of glutamatetoxicity, the pyramidal neuronsalso lost (10 min) glutamateexposure and was observed at all time points immunoreactivity with the monoclonal antibody. Since pyrami- measured.Furthermore, the inhibition of CaM kinase II activity dal cells comprise approximately 30% of the neuronal popula- was dependentupon both the stimulation of the NMDA subtype tion (Coulter et al., 1992) the resultant lossof immunoreactivity of glutamate-gatedchannels and the presenceof extracellular within these cells would explain the magnitude of loss of im- calcium, [Ca2+],. The results also confirm earlier reports of munoreactivity that was apparently greater than the loss of en- NMDA (Choi et al., 1988; Michaels and Rothman, 1990; Churn zyme activity. Thus, it is possiblethat CaM kinase II activity in et al., 1993) and [Ca*+],, dependence(Choi, 1987; Choi and the glutamate-sensitivepyramidal neuronsmay be reduced by Hartley, 1993) of glutamate-inducedneuronal cell death. In ad- almost 100% under cytotoxic conditions. Further study of the dition, the observationsthat conditions which result in delayed total neuronal population in which neuronal cells lose kinase neuronal cell death also result in inhibition of CaM kinase II immunoreactivity will be performed in future studies.Thus, ex- activity are consistent with the hypothesis that preservation of citotoxic glutamate exposure resulted in an analogousaltered CaM kinase II activity is important for neuronalviability. These immunoreactivity of CaM kinase II as observed in the whole observationsprovide evidence that inhibition of CaM kinase II animal ischemia model (Onodera et al., 1990; Churn et al., activity may be an important step in the cascadeof events that 1992b). This observation suggeststhat excitotoxic glutamate- ultimately result in delayed neuronalcell death. induced CaM kinase II inhibition occurred by similar mecha- Inhibition of CaM kinase II activity is an early and long- nismsas that observed in whole animal models of ischemia. lasting phenomenonin global forebrain ischemia(Churn et al., Knowledge of the pharmacology of glutamate induced neu- 1990a,b, 1992a;Zivin et al., 1991; Aronowski et al., 1993). Al- though proteolytic destruction of kinase subunitshas been sug- rotoxicity is important for our understandingof neurotoxicity gested in prolonged ischemia (Yamamoto et al., 1992), the in- that occurs in vivo. Previous reports have demonstratedthat se- hibition of CaM kinase II in transient ischemiamodels has been lective blockade of the NMDA receptor channelsduring gluta- shown to be due to a posttranslationalmodification of the en- mate exposure resulted in preservation of neuronal viability zyme which results in inhibition of enzymatic activity (Aron- (Choi et al., 1988; Michaels and Rothman, 1990; Churn et al., owski et al., 1992; Churn et al., 1992a). Since the inhibition 1993). In the present study, pharmacologic blockage of the observedafter glutamateexcitotoxicity was not due to enzymatic NMDA channel with MK-801, but not the AMPA glutamate degradationof the kinase, it is reasonableto speculatethat sim- receptor channels(CNQX) preservedCaM kinase II activity. In ilar mechanismsthat resulted in global forebrain-induced inhi- addition, specific NMDA agonistsresulted in a similarinhibition bition of CaM kinase II activity were active in glutamate exci- of CaM kinase II activity as did glutamate application. The re- totoxicity-induced CaM kinase II inhibition. Therefore, the sults support the hypothesis that excitotoxic activation of primary neuronalculture model describedin this report provides NMDA receptor channelsresults in inhibition of CaM kinase II a powerful tool to characterize the excitotoxic inhibition of CaM activity. The sameNMDA receptor activation results in a de- kinase II and excitotoxicity-induced neuronal cell death. layed neuronalcell death. Thus, excitotoxic NMDA receptor ac- Decreasedimmunoreactivity of CaM kinase II with a mono- tivation resultsin inhibition of CaM kinase II activity and de- clonal antibody directed towards the p subunit of the enzyme layed neuronalcell death. has been reported in whole animal ischemia (Churn et al., CaM kinase II is highly enrichedin neuronsand is especially 1992a,b). In the gerbil ischemiamodel, the decreasedimmuno- concentratedin the synaptic regions(Kennedy et al., 1983a;Gol- reactivity was not due to autophosphorylation (Churn et al., denring et al., 1984b; Kelly et al., 1984). Therefore, CaM kinase 1992b)or to destruction of the enzyme since when balancedfor II may regulate important neuronal processesinvolved in neu- subunit staining the decreasedimmunoreactivity was still ob- rotransmission.Activation of NMDA receptorshas been shown served (Churn et al., 1992a,b).In addition, loss of immunoreac- to activate CaM kinase II in a calcium-dependentmanner (Fu- tivity was observed only in neuronal populations that undergo kunaga et al., 1992). The NMDA-dependent kinase activation delayed neuronal cell death, such as the pyramidal neuronsof results in the production of a calcium-independentform of the t

(60 kDa) subunit of CaM kinase II (see Materials and Methods). A, Control cultures showing neuronal specific immunofluorescence with IC3-3D6. B, Glutamate-treated cultures demonstrating a loss of immunoreactivity with the anti-CaM kinase II antibody. C, Phase micrograph showing the sameneurons in A from control-treatedculture. I>, Phasemicrograph showing the sameneurons in B from glutamate-treatedculture. 3212 Churn et al. * NMDA Excitotoxicity and CaM Kinase II Inhibition kinase that may contribute to the increased neuronal excitability status epilepticus (Perlin et al., 1992) and (Churn et al., observed in LTI? Data from this study demonstrate that patho- 1990b, 1992a; Zivin et al., 1991; Aronowski et al., 1993). In logical stimulation of NMDA receptors resulted in inhibition of stroke, early and long term inhibition of CaM kinase II activity CaM kinase II activity that preceded calcium-dependent, delayed suggests that inhibition of this major neuronal enzyme may be neuronal death. Since no modulation of Ca’+-independent activ- important in the excitotoxic events which culminate in neuronal ity was observed following glutamate treatment, the present re- cell death. In addition, immunohistochemical studies showed port suggests that pathological neuronal excitation resulted in a that loss of CaM kinase II immunoreactivity occurred in the total inhibition of CaM kinase 11 activity. The chronic inhibition same neuronal population that was sensitive to ischemia (On- of CaM kinase II activity may underlie some of the NMDA- odera et al., 1990; Churn et al., 1992b). This report describes induced pathological mechanisms that result in delayed neuronal analogous findings of CaM kinase II inhibition to those observed death or other neuropathologies such as epilepsy and in whole animal models. The inhibition of CaM kinase II activity activity. observed in both ischemia and excitotoxic preparations indicates It has been demonstrated that glutamate toxicity is dependent that the cell culture model may be a powerful tool to decipher upon influx of extracellular calcium (Choi, 1987; Choi et al., the intracellular mechanisms and the role of CaM kinase II in- 1988; Michaels and Rothman, 1990; Choi and Hartley, 1993). hibition in modulating the excitotoxic events that result in de- To characterize whether influx of extracellular Ca?+ is occurring layed neuronal death. in our system, confocal microscopic quantitation of Indo-l flu- orescence was performed. Both glutamate and KCI exposures References resulted in significant elevation of [Ca?‘],. However, [Ca2+], re- Abele AE, Scholl. KP, Scholz WK, Miller RJ (1990) Excitotoxicity mained elevated following the removal of the glutamate stimulus induced by enhanced excitatory neurotransmission in cultured hip- but returned to baseline levels upon removal of KC1 solution. pocampal pyramidal neurons. Neuron 4:4 I 3-4 19. This prolonged calcium transient observed in glutamate-treated Abney ER, Bartlett PP, Raff MC (1981) Astrocytes, ependymal cells, neurons has been proposed to be a causal factor in glutamate- and develop on schedule in dissociated cell cultures of embryonic rat brain. Dev Biol 83:301-310. induced, delayed neuronal cell death (Choi, 1987; Connor et al., Amador M, Dani JA (199 I) Protein kinase inhibitor, H-7, directly af- 1988; Glaum et al., 1990). Calcium entry was site specific for fects N-methyl-o-aspartate receptor channels. Neurosci Len l24:25 I- mediating cell death, since increases in [Ca”], by NMDA re- 255. ceptor activation and not by depolarization-gated calcium chan- Aronowski J, Grotta JC, Waxham MN (1992) Ischemia-induced trans- nels causes delayed neuronal cell death, and extended neuronal location of Ca?+/calmodulin-dependent protein kinase II: possible role in neuronal damage. J Neurochem 58: 1743-1753. depolarization (Sombati et al., 1991, Coulter et al., 1992). The Aronowski J, Waxham MN, Grotta JC (1993) Neuronal protection and results of the present study demonstrated that the calcium-in- preservation of calcium/calmodulin-dependent protein kinase II and duced CaM kinase II inhibition was also site specific, since cal- protein kinase C activity by dextrorphan treatment in global ischemia. cium entry through NMDA receptor activation, but not voltage- J Cereb Flow Metab 13550-557. Babcock-Atkinson E, Norenberg MD, Norenberg LO, Neary JT (1989) gated calcium channels caused kinase inhibition. Thus, the Calcium/calmodulin-dependent protein kinase activity in primary as- increase in [Ca2+], induced by excitotoxic glutamate treatment trocyte cultures. 2: 112-l 18. may alter select Ca’+-regulated cellular processes modulated by Banker GA, Cowan WM (1977) Further observations on hippocampal NMDA receptor activation that are important for neuronal via- neurons in dispersed cell cultures. J Comp Neurol 187:469+94. bility. The role of the site specific increases in [Ca?+], in causing Baskys A, Bernstein NK, Barolet AW, Carlen PL (1990) NMDA and quisqualate reduce a Ca’+-dependent K ’ current by a protein kinase- the inhibition of CaM kinase II activity needs to be further elu- mediated mechanism. Neurosci Lett I1 2:76-8 I. cidated. Burke BE, DeLorenzo RJ (1981) Ca*+- and calmodulin-stimulated en- It has been shown that removal of calcium from the incuba- dogenous phosphorylation of neurotubulin. Proc Natl Acad Sci USA tion medium during toxic glutamate exposure prevented neuro- 78:991-99s. Caroni P, Carafoli E (1983) The regulation of the NA +-Ca” exchanger toxicity (Choi, 1987; Michaeis and Rothman, 1990). Therefore, of heart sarcolemma. Eur J Biochem 132:451+60. we tested whether removal of calcium from the extracellular Choi DW (1987) Ionic dependence of glutamate neurotoxicity. J Neu- medium would protect CaM kinase II activity. Removal of ex- rosci 7:369-379. tracellular calcium during glutamate exposure blocked the ex- Choi DW (1988) Calcium-mediated neurotoxicity: relationship to spe- citotoxic inhibition of CaM kinase II activity. The finding that cific channel types and role in ischemic damage. Trends Neurosci I I : 465-469. removal of extracellular calcium protects CaM kinase II activity Choi DW (I 992) Bench to bedside: the glutamate connection. Science parallels two other observations of neurotoxicity. Choi et al. 258:241-243. (1987) have shown that removal of extracellular calcium pre- Choi DW, Hartley DM (1993) Calcium and glutamate-induced cortical vents the calcium-dependent, delayed (calcium-dependent) neu- neuronal death. Res Pub1 Assoc Res NervMent Dis 71:23-34. Choi DW. Rothman SM (1990) The role of glutamate neurotoxicitv in ronal cell death. In addition, Coulter et al., (1992) has shown hypoxic-ischemic neuronal death. Annu R& Neurosci 13: I7 1-l g2. that removal of extracellular calcium will also prevent the in- Choi DW, Maulucci Gedde M, Kriegstein AR (1987) Glutamate neu- duction of END (Coulter et al., 1992). The observation that re- rotoxicity in cortical cell culture. J Neurosci 7:357-368. moval of extracellular calcium during excitotoxic glutamate ex- Choi DW, Koh JY, Peters S (1988) Pharmacology of glutamate neu- posure also protected CaM kinase II activity may lead to insight rotoxicity in cortical cell culture: attenuation by NMDA antagonists. J Neurosci 8: 185-196. into the intracellular events which control both the development Churn SB, Taft WC, Billingsley MS, Blair RE, DeLorenzo RJ (199Oa) of END and progress to eventual neuronal degeneration. Temperature modulation of ischemic neuronal death and inhibition of Modulation of CaM kinase II activity has been demonstrated calcium/calmodulin-dependent protein kinase 11in gerbils. Stroke 2 I : in models of neuronal plasticity such as LTP (Lisman, 1985; 1715-1721. Churn SB, Taft WC, DeLorenzo RJ (1990b) Effects of ischemia on Lisman and Goldring, 1988; Malenka et al., 1989; Silva et al., multifunctional calcium/calmodulin-dependent protein kinase type 11 1992a,b), kindling (Wasterlain and Farber, 1984; Goldenring et in the gerbil. Stroke 2l:IIIl 12-111116. al., 1986) and also in models of neuronal cell death such as Churn SB, Taft WC, Billingsley MS, Sankaran B, DeLorenzo RJ The Journal of Neuroscience, April 1995, f5(4) 3213

(1992a) Global forebrain &hernia induces a post-translational mod- modulin-dependent protein kinase from mammalian brain that phos- ification of multifunctional calcium- and calmodulin-dependent ki- phorylates synapsin I: partial purification and characterization. J Neu- nase II. J Neurochem 59:1221-1232. rosci 3:818-831. Churn SB, Yaghmai A, Povlishock J, Rafiq A, DeLorenzo RJ (1992b) Koh JY, Choi DW (1991) Selective blockade of non-NMDA receptors Global forebrain &hernia results in decreased immunoreactivity of does not block rapidly triggered glutamate-induced neuronal death. calcium/calmodulin-dependent protein kinase II. J Cereb Blood Flow Brain Res 548:318-321. Metab 12:784-793. Lai Y, Nairn AC, Greengard P (1986) Autophosphorylation reversibly Churn SB, Sombati S, Taft WC, DeLorenzo RJ (1993) Excitotoxicity regulates the calcium/calmodulin-dependent protein kinase II. Proc affects membrane potential and calmodulin kinase II activity in cul- Nat1 Acad Sci 83:4253-4257. tured rat cortical neurons. Stroke 24:271-278. Lisman J (1985) A mechanism for memory storage insensitive to mo- Connor JA, Tseng HY (1988) Measurement of intracellular Ca*+ in lecular turnover: a bistable autophosphorylating kinase. Proc Nat1 cerebellar Purkinje neurons in culture: resting distribution and re- Acad Sci USA 82:3055-3057. sponse to glutamate. Brain Res Bull 21:353-361. Lisman J, Goldring M (1988) Evaluation of a model of long-term Connor JA, Wadman WJ, Hockberger PE, Wong WKS (1988) Sus- memory based on the properties of the Ca*+/calmodulin-dependent tained dendritic gradients of Ca2+ induced by excitatory amino acids protein kinase. J Physiol (Lond) 83:187-197. in CA1 hippocampal neurons. Science 240:649-653. Llinas R, McGuinness TL, Leonard CS, Sugimori M, Greengard P Coulter DA, Sombati S, DeLorenzo RJ (1992) Electrophysiology of (1985) Intra-terminal injection of synapsin I or calciuticalmodulin- glutamate neurotoxicity in vitro: induction of a calcium-dependent dependent kinase II alters release at the squid giant extended neuronal depolarization. J Neurophysiol 68:362-373. synapse. Proc Nat1 Acad Sci USA 82:3035-3039. DeLorenzo RJ (198 1) The calmodulin hypothesis of neurotransmission. Machu TK, Firestone JA, Browning MD (1993) Ca*+/calmodulin-de- Cell Calcium 2:365-385. pendent protein kinase II and protein kinase C phosphorylate a syn- DeLorenzo RJ (1983) Calcium-calmodulin protein phosphorylation in thetic peptide corresponding to a sequence that is specific for the neuronal transmission: a molecular approach to neuronal excitability gamma 2L subunit of the GABA, receptor. J Neurochem 61:375- and anticonvulsant drug action. Adv Neurol 34:325-338. 377. DeLorenzo RJ, Gonzalez B, Goldenring J, Bowling A, Jacobson R Malenka RC, Kauer JA, Perkel DJ, Mauk MD, Kelly PT, Nicoll RA, (1982) Ca*+-calmodulin tubulin kinase system and its role in medi- Waxham MN (1989) An essential role for postsynaptic calmodulin ating the Ca*+ signal in brain. Prog Brain Res 56:257-286. and protein kinase activity in long-term potentiation. Nature 340: Erondu NE, Kennedy MB (1985) Regional distribution of type II cal- 554-551. cium/calmodulin-dependent protein kinase in rat brain. J Neurosci McGlade-McGulloh E, Yamamoto H, Tan S-E, Brickey D, Soderling 5:3270-3211. TR (1993) Phosphorylation and regulation of glutamate receptors by Fukunaga K, Soderling TR, Miyamoto E (1992) Activation of Ca2+/ calcium/calmodulin-dependent protein kinase II. Nature 362:640- calmodulin-dependent protein kinase II and protein kinase C by glu- 642. tamate in cultured rat hippocampal neurons. J Biol Chem 267:22527- Michaels RL, Rothman SM (1990) Glutamate neurotoxicity in vitro: 22533. antagonist pharmacology and intracellular calcium concentrations. J Glaum SR, Scholz WK, Miller RJ (1990) Acute- and long-term glu- Neurosci 10:283-292. tamate-mediated regulation of [Ca++], in rat hippocampal pyramidal Murphy SN, Miller RJ (1988) A glutamate receptor regulates CaZ+ neurons in vitro. J Pharmacol Exp Ther 253:1293-1302. mobilization in hiuuocamual neurons. Proc Nat1 Acad Sci USA 85: Goldenring JR, Gonzalez B, McGuire JS Jr, DeLorenzo RJ (1983) Pu- 8737-8741. __ I rification and characterization of a calmodulin-dependent kinase from Onodera H, Hara H, Kogure K, Fukunaga K, Ohta Y, Miyamoto E rat brain cytosol able to phosphorylate tubulin and microtubule-as- (1990) Ca2+/calmodulin-dependent protein kinase II immunoreactiv- sociated proteins. J Biol Chem 258: 12632-12640. ity in the rat hippocampus after forebrain ischemia. Neurosci Lett Goldenring JR, Casanova JE, DeLorenzo RJ (1984a) Tubulin-associ- 113:134-138. ated calmodulin-dependent kinase: evidence for an endogenous com- Ouimet CC, McGuiness TL, Greengard P (1984) Immunocytochemical plex of tubulin with a calcium-calmodulin-dependent kinase. J Neu- localization of calcium/calmodulin-dependent kinase II in rat brain. rochem 43:1669-1679. Proc Nat1 Acad Sci USA 81:5604-5608. Goldenring JR, McGuire JS Jr, DeLorenzo RJ (1984b) Identification Patton BL, Molloy SS, Kennedy MB (1993) Autophosphorylation of of the major postsynaptic density protein as homologous with the type II CaM kinase in hippocampal neurons: localization of phospho- major calmodulin-binding subunit of a calmodulin-dependent protein and dephosphokinase with complementary phosphorylation site-spe- kinase. J Neurochem 42:1077-1084. cific antibodies. Mol Biol Cell 4:159-172. Goldenring JR, Wasterlain CG, Oestreicher AB, de Graan PN, Farber Perlin JB, Churn SB, Lothman EW, DeLorenzo RJ (1992) Loss of type DB, Glaser G, DeLorenzo RJ (1986) Kindling induces a long-lasting II calcium/calmodulin-dependent kinase activity correlates with change in the activity of a hippocampal membrane calmodulin-de- stages of development of electrographic in status epilepticus pendent protein kinase system. Brain Res 377:47-53. in rat. Epilepsy Res 11: 11 l-l 18. Hanson PI, Schulman H (1992) Inhibitory autophosphorylation of mul- Portzehl H, Caldwell PC, Reugg JC (1964) The dependence of con- tifunctional Ca*+/calmodulin-dependent protein kinase analyzed by traction and relaxation of muscle fibers from the crab Muia squimndo site-directed mutagenesis. J Biol Chem 267:17216-17224. on the internal concentration of free calcium ions. Biochim Biophys Hartley DM, Kurth MC, Bjerkness L, Weiss JH, Choi DW (1993) Glu- Acta 79:581-591. tamate receptor-induced 45Caz+ accumulation in cortical cell culture Rothman SM (1986) Glutamate and anoxic neuronal death in vitro. correlates with subsequent neuronal degeneration. J Neurosci 13: Adv Exp Med Biol 203:687-695. 199332000. Sakakibara M, Alkon DL, DeLorenzo RJ, Goldenring JR, Neary JT, Hashimoto Y, Soderling TR (1987) Calcium calmodulin-dependent Heldman E (1986) Modulation of calcium-mediated inactivation of protein kinase II and calcium phospholipid-dependent protein kinase ionic currents by calcium/calmodulin-dependent protein kinase II. activities in rat tissues assayed with a synthetic peptide. Arch Bio- Biophys J 50:319-327. them Biophys 252:418-425. Schulman H (1989) The multifunctional calcium/calmodulin-depen- Jameson L, Frey T, Zeeberg B, Dalldorf F, Caplow M (1980) Inhibition dent protein kinase. In: Advances in second messenger and phospho- of microtubule assembly by phosphorylation of microtubule-associ- protein research (Greengard P, Robinson GA, eds), pp 39-112. New ated proteins. Biochemistry 19:2472-2479. York: Raven. Kelly PT, McGuinness TL, Greengard P (1984) Evidence that the major Shields SM, Ingebritsen TS, Kelly PT (1985) Identification of protein post synaptic density protein is a component of calcium/calmodulin phosphatase 1 in synaptic junctions: dephosphorylation of endoge- protein kinase. Proc Nat1 Acad Sci USA 81:945-949. nous calmodulin-dependent kinase and synapse-enriched phospho- Kennedy MB, Bennett MK, Erondu NE (1983a) Biochemical and im- proteins. J Neurosci 5:3414-3422. munochemical evidence that the “major postsynaptic density pro- Silva AJ, Paylor R, Wehner JM, Tonegawa S (1992a) Impaired spatial tein” is a subunit of a calmodulin-dependent protein kinase. Proc learning in alpha-calcium-calmodulin kinase II mutant mice [see Nat1 Acad Sci USA 80:7357-7361, comments]. Science 257:206-211. Kennedy MB, McGuinness T, Greengard P (1983b) A calcium/cal- Silva AJ, Stevens CF, Tonegawa S, Wang Y (1992b) Deficient hippo- 3214 Churn et al. * NMDA Excitotoxicity and CaM Kinase II Inhibition

campal long-term potentiation in alpha-calcium-calmodulin kinase II dulin-dependent phosphorylation of synaptic plasma membrane pro- mutant mice [see comments]. Science 257:201-206. teins in rat hiooocamuus. Proc Nat1 Acad Sci USA 81:1253-1257. Sombati S, Coulter DA, DeLorenzo RJ (1991) Neurotoxic activation Yamamoto H, Fukunag’a K, Goto S, Tanaka E, Miyamoto E (1985) of glutamate receptors induces an extended neuronal depolarization Calcium, calmodulin-dependent regulation of microtubule formation in cultured hiooocamnal neurons. Brain Res 566:3 16-3 19. via phosphorylation of microtubule-associated protein 2, tau factor, Taft WC, Tennes kees KA, Blair RE, Clifton GL, DeLorenzo RJ (1988) and tubulin and comparison with the cyclic AMP-dependent phos- Cerebral ischemia decreases endogenous calcium-dependent protein phorylation. J Neurochem 44:759-768. phosphorylation in gerbil brain. Brain Res 447:159-163. Yamamoto H, Fukunaga K, Lee K, Soderling TR (1992) Ischemia- Wadman WJ, Connor JA (1992) Persisting modification of dendritic induced loss of brain calciuticalmodulin-dependent protein kinase calcium by excitatory amino acid stimulation in isolated CA1 neu- II. J Neurochem 581110-l 117. rons. Neuroscience 48293-305. Zivin JA, Kochhar A, Saitoh T (1991) Protein phosphorylation during Wasterlain CG, Farber DB (1984) Kindling alters the calciuticalmo- ischemia. Stroke [Suppl] 21 :I11117-111121.