• Tidak ada hasil yang ditemukan

Directory UMM :Data Elmu:jurnal:B:Biological Psichatry:Vol49.Issue1.2001:

N/A
N/A
Protected

Academic year: 2017

Membagikan "Directory UMM :Data Elmu:jurnal:B:Biological Psichatry:Vol49.Issue1.2001:"

Copied!
12
0
0

Teks penuh

(1)

Antipsychotic Treatment Induces Alterations in

Dendrite- and Spine-Associated Proteins in

Dopamine-Rich Areas of the Primate Cerebral Cortex

Michael S. Lidow, Zan-Min Song, Stacy A. Castner, Patrick B. Allen,

Paul Greengard, and Patricia S. Goldman-Rakic

Background:

Mounting evidence indicates that long-term

treatment with antipsychotic medications can alter the

morphology and connectivity of cellular processes in the

cerebral cortex. The cytoskeleton plays an essential role in

the maintenance of cellular morphology and is subject to

regulation by intracellular pathways associated with

neu-rotransmitter receptors targeted by antipsychotic drugs.

Methods:

We have examined whether chronic treatment

with the antipsychotic drug haloperidol interferes with

phosphorylation state and tissue levels of a major

den-dritic cytoskeleton–stabilizing agent,

microtubule-associ-ated protein 2 (MAP2), as well as levels of the dendritic

spine–associated protein spinophilin and the synaptic

vesicle–associated protein synaptophysin in various

re-gions of the cerebral cortex of rhesus monkeys.

Results:

Among the cortical areas examined, the

prefron-tal, orbiprefron-tal, cingulate, motor, and entorhinal cortices

displayed significant decreases in levels of spinophilin,

and with the exception of the motor cortex, each of these

regions also exhibited increases in the phosphorylation of

MAP2. No changes were observed in either spinophilin

levels or MAP2 phosphorylation in the primary visual

cortex. Also, no statistically significant changes were

found in tissue levels of MAP2 or synaptophysin in any of

the cortical regions examined.

Conclusions:

Our findings demonstrate that long-term

haloperidol exposure alters neuronal cytoskeleton– and

spine–associated proteins, particularly in dopamine-rich

regions of the primate cerebral cortex, many of which

have been implicated in the psychopathology of

schizo-phrenia. The ability of haloperidol to regulate cytoskeletal

proteins should be considered in evaluating the

mecha-nisms of both its palliative actions and its side effects.

Biol Psychiatry 2001;49:1–12 ©

2001 Society of

Biolog-ical Psychiatry

Key Words:

Antipsychotic drugs, dendrite, spine,

syn-apse, microtubule-associated protein 2, spinophilin,

synaptophysin

Introduction

S

ince the introduction of antipsychotic medications

nearly 40 years ago, the main emphasis of studies on

the effects of these drugs in brain tissue has been on

alterations in neurotransmitters and their receptors (for a

review, see Csernansky 1996). There is, however,

mount-ing evidence that antipsychotics may produce

morpholog-ical changes in cellular elements in several regions of the

brain, particularly in the association areas of the cerebral

cortex (Benes et al 1985; Klinzova et al 1989, 1990;

Meshul et al 1992; Uranova et al 1991; Vincent et al 1991,

1994). It has even been suggested that these

slow-devel-oping morphological changes might explain why the

pharmacologic treatment of psychosis typically requires

several weeks to attain its full effect and why it may take

months to reverse this effect after cessation of treatment

(Benes et al 1985). It has also been proposed that the

antipsychotic-induced morphological alterations may

un-derlie the side effects produced by these drugs (Kelley et

al 1997; Seeman 1988). Interest in the effects of

antipsy-chotic medications on the morphology of cortical cells is

further reinforced by the discovery of alterations in the

volume and organization of the neuropil in postmortem

cortical tissue from schizophrenic patients (Anders 1978;

Garey et al 1998; Glantz and Lewis 1997; Selemon et al

1996; Uranova et al 1996), raising the question as to

whether these changes are associated with the disease

itself or its treatment.

The ability of antipsychotic drugs to affect cell

mor-phology can be predicted from their binding to

dopami-nergic and other neurotransmitter receptors (Seeman

1990), which are coupled to second messengers that

regulate the activity of kinases and phosphatases

(Keba-bian and Greengard 1971; Roth et al 1998; Walsh et al

1972; Yurko-Mauro and Friedman 1995). These enzymes

control the phosphorylation states of such proteins as

From the Department of Oral and Craniofacial Biological Sciences, University of

Maryland, Baltimore (MSL, Z-MS), Section of Neurobiology, Yale University School of Medicine, New Haven, Connecticut (SAC, PSG-R), and Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York (PBA, PG).

Address reprint requests to Michael S. Lidow, Ph.D., University of Maryland, Baltimore, OCBS Department, Room 5-A-12, HHH, 666 W. Baltimore Street, Baltimore MD 21201.

Received June 15, 2000; revised August 25, 2000; accepted August 28, 2000.

© 2001 Society of Biological Psychiatry 0006-3223/01/$20.00

(2)

microtubule-associated protein 2 (MAP2; Goldenring et al

1985; Sloboda et al 1975; Tsuyama et al 1986; Walaas and

Nairn 1989). MAP2 contains multiple phosphorylatable

residues, and the levels of phosphorylation of this protein

are inversely proportional to its ability to stabilize

den-dritic microtubules (Cleveland and Hoffman 1991;

Mac-cioni and Cambiazo 1995; Tsuyama et al 1986, 1987;

Wiche et al 1991). As microtubules are among the major

cytoskeletal constituents involved in the maintenance of

dendritic processes (Cleveland and Hoffman 1991; Keith

1990; Shea and Beermann 1994; Yamada et al 1970), any

changes in their stability could affect major cellular

compartments of neurons—their dendrites, spines, and

synapses. Despite the obvious importance of

understand-ing the effects of antipsychotic drugs on the cytoskeleton

of cortical cells, their effects on the phosphorylation of

MAP2 and other cytoskeletal proteins in the cortex are not

known.

During the past several years our laboratories have been

involved in an analysis of the effects of chronic

antipsy-chotic treatment on the integrity of the primate cortex. We

have previously reported a significant upregulation of D

2

receptors and downregulation of D

1

receptors in the

prefrontal and temporal cortical regions of the rhesus

monkey brain following chronic treatment with several

typical and atypical antipsychotic drugs (Lidow et al 1997;

Lidow and Goldman-Rakic 1994). The cortex of these

monkeys also exhibits increased glial density (Selemon et

al 1999), although this increase was not induced by all of

the antipsychotic drugs examined. In particular,

haloperi-dol was not among the drugs that induced significant

gliosis.

This study extends our investigations to the influence of

chronic treatment with antipsychotic drugs—in this

in-stance, haloperidol— on tissue levels and degree of

phos-phorylation of the dendritic cytoskeleton–stabilizing

pro-tein MAP2 in multiple regions of the monkey cerebral

cortex. In addition, we examined the impact of this

treatment on cortical levels of spinophilin, the dendritic

spine–affiliated protein that has been implicated in linking

plasma membrane–associated synaptic components, such

as dopamine D

1

and D

2

receptors, to the actin cytoskeleton

(Allen et al 1997; Smith et al 1999; Yan et al 1999). We

also evaluated cortical levels of synaptophysin, a synaptic

marker commonly associated with synaptic vesicles

(East-wood et al 1995; Masliah et al 1990) presumed to act as an

exocytotic fusion pore (Bajjalieh and Scheller 1995;

Edel-mann et al 1995). Effects of haloperidol treatment on

spinophilin and synaptophysin were examined because

alterations in the density of spines and synapses containing

these proteins have previously been noted in the cortex of

haloperidol-exposed animals (Benes et al 1985; Klinzova

et al 1989, 1990; Meshul et al 1992; Vincent et al 1991).

In this investigation we studied monkeys that were 15

years of age or older to model the effects of drug treatment

on brain cells in a population of older patients. A parallel

study is currently in progress on a younger cohort of

monkeys. We report the findings on the older animals now

because of the insight they provide for neural

vulnerabil-ities in aged individuals as well as for possible

implica-tions for chronic medication in clinical practice.

Methods and Materials

Animals

This study included four haloperidol-treated and four drug-naive control female rhesus monkeys (Macaca mulatta). The monkeys in the drug-exposed group were 15, 19, 19, and 20 years of age, whereas the animals in the control group were 15, 16, 19, and 25 years of age. The animals were kept in individual cages in accordance with Yale Animal Use and Care Committee guide-lines for nonhuman primates. All monkeys were fed High Protein Monkey Chow (Ralston Purina, Saint Louis), were given fruit twice a day, and had fresh water available ad libitum. The animals were also provided with standard enrichment devices: logs, dog toys, plastic chains, and mirrors.

Drug Treatment and Tissue Collection

Haloperidol (in powder form) was obtained from RBI (Natick, MA). For administration, a stock solution of haloperidol:sucrose (1:50) was prepared, from which daily doses were portioned out and given to animals within fruit treats such as pieces of banana, prune, apple, or marshmallow. The animals received the drug twice a day for a period of 1 year. During the first month of treatment the daily dose was 0.07 mg/kg. During the second month it was increased to 0.14 mg/kg. After that, the daily dose was increased to 0.20 and 0.27 and finally to 0.35 mg/kg at 2-week intervals. The latter dose was maintained throughout the rest of the treatment period. For one monkey, the final daily dose was 0.42 mg/kg, which was dictated by requirements of testing for cognitive impairments, also conducted in these animals. The final doses of haloperidol employed in this study fall within the therapeutic range given to psychiatric patients during mainte-nance treatment (Physicians’ Desk Reference 1999). The control animals received fruit treats only. During the entire period of treatment the animals displayed no signs of extrapyramidal side effects.

Between 12 and 18 hours after the last treatment, the animals were anesthetized with sodium pentobarbital. Their brains were rapidly removed, dissected, and immersed in liquid nitrogen for storage.

(3)

animals at the time of perfusion. Also, since the estrous cycles in rhesus monkeys display seasonal variations (Hutz et al 1985; Walker et al 1983), all the animals were perfused in the period from November through February, which is within the breeding season for these species.

Samples from the following cortical areas were analyzed: frontal pole (cortical area 10; Walker 1940), dorsolateral pre-frontal cortex (area 46; Walker 1940), dorsomedial prepre-frontal cortex (area 9; Walker 1940), anterior orbital cortex (area 11; Walker 1940), posterior orbital cortex (area 13; Walker 1940), anterior cingulate cortex (area 24; Walker 1940), prelimbic cortex (area 25; Walker 1940), premotor cortex (lateral portion of area 6; Brodmann 1994), primary motor cortex (area 4; Brod-mann 1994), primary visual cortex (area 17; BrodBrod-mann 1994), and entorhinal cortex (area 28; Brodmann 1994). The cortical regions examined are shown in Figure 1. Samples of all brain regions were collected from both hemispheres, but those from each hemisphere were processed separately.

Tissue Levels of MAP2, Spinophilin, and

Synaptophysin

TISSUE HOMOGENATES. Tissue samples were

homoge-nized for 3 min on a TriR Homogenizer (Cole-Palmer, Vernon Hill, IL) in 100 volumes of ice-cold TBS buffer (0.5 mol/L Tris base [pH 8.0] containing 0.1 mol/L NaCl and 0.8 mmol/L phenymethylsulfonyl fluoride).

SLOT BLOTS FOR PROTEIN ANALYSIS. Comparative lev-els of specific proteins in the sample homogenates were exam-ined using slot blots on NitroPure membranes (Osmonics, Westbrough, MA) prepared with Bio-Dot SF Microfiltration Apparatus (Bio-Rad, Hercules, CA). Before the blotting, each tissue homogenate was diluted 1:100 with the TBS buffer, and 200mL of the resultant solution were used per blot.

For immunolabeling, membranes were preincubated for 1 hour at room temperature in blocking solution containing 5% dry milk and 0.2% Tween, in PBS buffer (137 mmol/L NaCl, 2.7 mmol/L KCl, 4.3 mmol/L Na2HPO4, and 1.4 mmol/L KH2PO4; pH 7.3).

Incubation with protein-specific antibodies diluted in the same buffer was conducted overnight at 4°C. The dilution of MAP2 and spinophilin antibodies was 1:2500; for synaptophysin anti-bodies, the dilution was 1:1000. After incubation, the membranes were washed 235 min in PBS buffer and exposed for 1.5 hours to the secondary peroxidase-conjugated antibodies diluted 1:62500 in the blocking solution described above. Visualization of labeling was conducted with the Super Signal Chemilumines-cence Substrate (Pierce, Rockford, IL). The images were pro-duced by opposing transparent plastic-wrapped chemilumines-cence-soaked membranes to an X-Omat AR Film (Kodak, Rochester, NY) for a period of 2–25 min. For analysis with Universal Software (Advanced American Biotechnology, Fuller-ton, CA), the film images of slot blots were digitized on a UC 1260 flat bed scanner (U-Max, Hsinchy, Taiwan). For examina-tion of the levels of each specific protein, the tissue samples from the same brain region of the same hemisphere of all eight animals used in this study were always processed simultaneously on a single membrane and blots for every sample were done in

(4)

controls for linearity, represented by six blots generated by a serial dilution of monkey brain prepared for this purpose (Figure 2B). The film images of the sample blots from a membrane were accepted for analysis only if all of them had gray value within the range of those produced by the controls for linearity and if the relationship between the gray values of the images of the

linearity control blots and the tissue dilution in these blots was in the linear range. In addition, every membrane included a line of six blots produced by bovine serum albumin (BSA), prepared as the tissue samples, to check the level of background immunola-beling. The BSA blots produced no detectable images on any of the films examined in this study (Figure 2B).

ANTIBODIES. Specific proteins were labeled with the

fol-lowing antibodies: monoclonal antibodies to the high–molecular weight a and b isoforms of MAP2 (Sigma, Saint Louis), the rabbit polyclonal antibodies RU 144 to spinophilin (Allen et al 1997), and the mouse monoclonal antibodies EP10 to synapto-physin (StressGen Biotechnology, Victoria, Canada). The sec-ondary goat antimouse and goat–antirabbit peroxidase– conju-gated antibodies were purchased from Jackson Immunoresearch (West Grove, PA). The specificity of the primary antibodies was tested on Western blots of monkey cortical and striatal tissues. The homogenates were mixed 5:1, with the loading buffer containing1.0 mol/L Tris-HCl (pH 6.8), 20% sodium dodecyl sulfate (SDS), 50% glycerol, 0.2% Bromophenol blue, and 4.9% DTT. Thirty microliters of this mixture were loaded per well in Ready-made 4 –15% Gradient SDS Gels (Bio-Rad). The gels were run for 1.5 hours at 100 V using a Bio-Rad Ready Gel Cell with Bio-Rad Tris/Glycine/SDS Running Buffer. The transfer on a PVDF-Plus membrane (Osmonics) was performed at 100 A, overnight at 4°C also using a Bio-Rad Ready Gel Cell with Bio-Rad Tris/Glycine Buffer. Immunolabeling of the membranes was performed as described above for the slot blots. The specificity of all the antibodies used in this test was demonstrated by the observation that they produced labeling only in bands with molecular weight in the appropriate range (Figure 2A): MAP2; 280 kd (two bands of close molecular weight that representeda

andbisoforms of this protein were visualized; Johnson and Jope 1992), spinophilin;140kD (Allen et al 1997), and synaptophy-sin;38 kD (Honer et al 1992).

SLOT BLOTS FOR ANALYSIS OF THE DNA LEVELS. The

slot blots were used for measurement of the comparative levels of DNA in the sample homogenates. Similar to the blots for the protein analysis, the blots for analysis of DNA were prepared using a Bio-Dot SF Microfiltration Apparatus (Bio-Rad, Her-cules, CA). However, here it was loaded with the Hybond-N Nylon membrane (Amersham, Piscataway, NJ). Also, before blotting, the tissue homogenates were diluted 1:20 in 90 mmol/L citrate buffer (pH 7.0) containing 0.9 mol/L NaCl, 3.0 mmol/L EDTA, and 40mg/mL RNase A. For visualization of the DNA, the dried membranes were soaked for 15 min in SYBR DX DNA-specific Blot Stain (Molecular Probe, Eugene, OR) mixed 1:1000 with 89 mmol/L Tris-H3BO3buffer (pH 8.0) containing

10 mmol/L EDTA. The staining was observed with the UV Photo Viewer Illumination System (Ultra-Lum, Paramount, CA) at the wavelength of 454 nm. The system also digitized the images for densitometric analysis with Universal Software. A typical exam-ple of the digitized image of a membrane stained for DNA is presented in Figure 2C. As in the case of the blots for the protein analysis, samples from the same region of the same hemisphere were processed simultaneously for all eight animals, with blots for every sample done in triplicate. The controls for linearity Figure 2. Representative examples of the images of the Western

(5)

consisted of slot blots of six serial dilutions of herring sperm DNA (Figure 2C). The background staining on each membrane was checked with six blots of BSA prepared in a manner identical to that of the experimental samples (Figure 2C). On all membranes used for densitometric analysis, the BSA blots generated no staining.

DETERMINATION OF PROPORTION OF NEURONS IN THE TISSUE. Since the overwhelming majority of cells in the brain

are not in a process mitosis and have a single complement of identical DNA, it is reasonable to expect that the proportion of the neuronal DNA in a blot would be very close to the proportion of the neuronal cells in the tissue sample. Therefore, it was of interest to determine the proportions of the neuronal DNA in the samples collected for this study. For this purpose, three randomly cut slabs from each tissue sample were processed for direct three-dimensional counting of cell nuclei. The counting was performed as outlined in Selemon et al (1996, 1999) using a Macintosh-based computer system described in Williams and Rakic (1988b). For counting, the slabs were sliced into 80-mm sections on an HM 500 OM cryostat (Zeiss, Walldorf, Germany), and the sections were stained with cresyl violet. The counting was conducted on one section per slab. For each section, the counting was performed in two nonoverlapping randomly se-lected counting boxes (55mm wide325mm deep) stretching across the brain structure. Therefore, we examined six counting boxes for every tissue sample (two boxes 3 three separate sections). Neuronal nuclei were identified based on the criteria of Williams and Rakic (1988a). The proportions of neuronal nuclei among all the nuclei were calculated for every counting box. These data were then used for calculation of the mean propor-tions6SEMs of neuronal nuclei for every brain region of each animal.

MEASUREMENT OF TOTAL PROTEIN. The measurement of total protein in the sample tissue homogenates was performed with a Modified Lowry Protein Assay Kit (Pierce) on a Hitachi U110 spectrophotometer (Hitachi USA, San Jose, CA).

EXPRESSION OF THE DATA AND STATISTICAL ANALY-SIS. MAP2 and spinophilin are produced by cortical neurons, and synaptophysin, although situated presynaptically, still repre-sents the synapses on cortical cells. Therefore, we wanted to evaluate the effects of the drug treatment on the levels of MAP2, spinophilin, and synaptophysin per neuron in every brain region examined. Since it is not possible to determine precisely the number of cells in the tissue samples used for blots, the closest representation of the quantity of a specific protein per neuron is to divide the amount of this protein by the DNA amount in the same sample and multiply by the proportion of neuronal cells in the sample tissue. In practice, the mean gray values of the images generated by the protein-specific immunostaining of the blots from each sample were divided by the mean gray value of the images generated by the DNA staining of the blots from this sample and multiplied by the mean proportion of the neurons in the counting boxes in the sections also obtained from the same sample. In addition, we expressed the levels of all specific

proteins examined (gray value of blot labeling) per unit of total protein in the sample.

While 22 cortical areas (11 areas per hemisphere) were examined in this study, the goal of the experiments did not include any comparison of protein levels between these areas. The basic analysis of the data, therefore, was a two-tailedttest comparing the levels of each protein in individual cortical area between control and treated animals. Given that four different proteins were measured in each cortical area, however, we adopted a conservative approach by applying a Bonferroni correction to thepvalues calculated from thettests. Hence, the

pvalue used in determining the statistical significance of each analysis was equal to the p value calculated from the t test divided by 4, which is the number of proteins analyzed in each cortical area.

Analysis of Phosphorylation of MAP2

PROCESSING OF THE TISSUE. The analysis of the effect of

(6)

(San Francisco). The antibodies for immunolabeling of MAP2 were the same as those used for immunoprecipitation (dilution 1:2500). In this case, the secondary antibodies visualized both the immunolabeling and immunoprecipitating antibodies. How-ever, the immunolabeling of MAP2 can be easily distinguished because the molecular weights ofaandbisoforms of MAP2 are in the range of 280 kd, whereas the molecular weights of the heavy and light chains of the immunoprecipitating antibodies are 50 kd and 25 kd, respectively (Alberts et al 1995). Rabbit polyclonal antibodies for phosphoserine, phosphothreonine, and phosphotyrosine were purchased from Zymed Laboratories and used at a dilution 1:500. The peroxidase-conjugated secondary antibodies were the same as those used in the immunolabeling of the slot blots. Their dilution was 1:62500. A typical example of the digitized film images of the immunoblots generated in this part of the study is presented in Figure 3. All Western blots were generated in six replicates. Also, each membrane processed for immunolabeling contained controls for linearity that, as de-scribed earlier for the slot blots, were used to ensure that the gray values obtained in the film images were in the linear range. The controls for linearity were made of six serial dilutions of the monkey brain tissue collected and stored for this purpose. The controls for linearity were placed manually, as 2-mL dot blots, on the edge of the membrane immediately upon its removal from the transfer apparatus, before the drying step (Figure 3).

EXPRESSION OF THE DATA AND STATISTICAL ANALY-SIS. The residue-specific phosphorylation in every blot of a

given sample was expressed as the ratio of the gray value of the film images produced by the phosphoaminoacid immunolabeling and the gray value of the images produced by the immunolabel-ing of the immunoprecipitated protein of this blot. For every set of blots on a single membrane, the ratios obtained were then normalized to the ratio generated by the sample from the 15-year-old control animal. Since every assay was performed in six replicates (on six separate membranes), the normalized ratios from the matching samples in all six replicates (membranes) were averaged for statistical analysis. The data obtained in this way were appropriate only for comparison of the levels of residue-specific MAP2 phosphorylation in the same region of the same hemisphere of control and experimental animals. The statistic employed for this purpose was a two-tailed ttest. In assessing statistical significance, we used the Bonferroni correc-tion with pvalue divided by 3, the number of specific assays performed in each brain area.

Results

Proportion of Neuronal Cells in Multiple Cortical

Regions

Our calculations of the proportions of neuronal nuclei

among the cell nuclei in the samples from all the cortical

regions examined in this study showed that neurons

constitute approximately half of the cells in these regions

(Table 1). There were no statistical differences between

the proportions of neuronal nuclei in the

haloperidol-treated and control animals in any of the cortical regions in

either hemisphere (Table 1).

Levels of Spinophilin, MAP2, and Synaptophysin

Comparative analysis of spinophilin levels in the

haloperidol-treated and control animals showed that the drug induced a

statistically significant downregulation of this protein in all of

the frontal and temporal cortical areas examined in both

hemispheres (Figure 4). These areas included the cortex of

the frontal pole, dorsolateral and dorsomedial prefrontal

cortices, anterior and posterior orbital cortices, anterior

cin-gulate and prelimbic cortices, premotor and primary motor

cortices, and entorhinal cortex. Among the cortical areas

Table 1. Percentages6SEMs of Neurons in 11 Different Cortical Regions of the Right Hemisphere of the Long-Term Haloperidol-Treated and Drug-Naive Control Monkeys

Frontal pole

Dorsolateral prefrontal

Dorsomedial prefrontal

Anterior orbital

Posterior orbital

Anterior

cingulate Prelimbic Premotor

Primary motor

Primary

visual Entorhinal

Control animals 57.464.2 54.864.6 53.062.2 57.064.0 56.463.0 49.965.6 52.662.2 49.764.0 55.463.8 60.964.2 58.364.6 Haloperidol-treated

animals

55.865.2 56.163.2 52.363.6 55.863.6 55.464.4 51.163.6 52.661.4 49.063.4 54.662.0 59.365.0 56.966.0

ttest p..05 p..05 p..05 p..05 p..05 p..05 p..05 p..05 p..05 p..05 p..05 Figure 3. Film images of Western blots of a set of MAP2

(7)

analyzed in this study, only the primary visual cortex did not

display a statistically significant decline in this protein

(Fig-ure 4). The same results were obtained whether the levels of

spinophilin were expressed per neuronal DNA or per total

protein (Figure 4).

We also found that the haloperidol-treated monkeys had

elevated levels of MAP2 in all cortical regions examined

in both hemispheres, whether the levels of this protein

were calculated per neuronal DNA or per total protein of

the sample (Figure 4). However, this upregulation did not

reach statistical significance in any region (Figure 4).

We detected no statistically significant differences in

the levels of synaptophysin between the drug-treated and

control animals in any cortical region examined (Figure 4).

Phosphorylation of MAP2

The analysis of MAP2 phosphorylation in the

haloperidol-treated and control animals revealed a statistically

signif-icant drug-induced increase in the levels of

phosphoryla-tion of this protein on the serine residue in the cortex of the

frontal pole, dorsolateral and dorsomedial prefrontal

cor-tices, anterior and posterior orbital corcor-tices, anterior

cin-gulate and prelimbic cortices, and entorhinal cortex

(Fig-ure 5). Increases in MAP2 serine phosphorylation were

also observed in the premotor, primary motor, and primary

visual cortical areas, but these increases did not reach

statistical significance (Figure 5). Increases in the levels of

MAP2 phosphorylation were also observed on the

threo-nine residue in all brain regions, but these increases were

also not statistically significant (Figure 5). No significant

effects on the tyrosine residue phosphorylation of MAP2

were found in the haloperidol-treated animals (Figure 5).

Discussion

Selective Regional Vulnerability of MAP2 and

Spinophilin to Chronic Haloperidol Treatment

This study demonstrates that chronic haloperidol treatment

significantly increases MAP2 phosphorylation on the

serine residue and significantly downregulates spinophilin

in specific regions of the primate cerebral cortex.

Further-more, the haloperidol-induced downregulation of

spi-nophilin is detectable independent of whether the levels of

this protein are expressed per neuronal DNA or per total

protein in the tissue. This indicates that haloperidol

treat-ment selectively affected spinophilin levels, without

inter-fering with the proportion of neurons in the cortex, as

demonstrated by the cell counts conducted in this study,

and without altering the total protein content of the cortical

cells. The protein specificity of the detected effects is also

emphasized by the absence of statistically significant

(8)

changes in the levels of MAP2 and synaptophysin in the

haloperidol-treated animals.

A central observation of this study is that the effects of

haloperidol were detectable in such cortical areas as the

frontal pole; the prefrontal, anterior cingulate, prelimbic,

and entorhinal regions; and the motor and premotor

cortices, all of which are known to contain extensive

dopaminergic innervation (Berger et al 1988;

Nieuwen-huys 1985; Williams and Goldman-Rakic 1993). These

areas, except the motor cortex, also contain the highest

densities of D

2

receptors in the monkey cerebral cortex

(Lidow et al 1998). Conversely, we detected no significant

haloperidol-induced alterations in the aforementioned

pro-teins in the visual cortex, which has a barely detectable

dopamine innervation and is D

2

receptor poor (Berger et al

1988; Lidow et al 1989). Interestingly, in the motor cortex,

which is richly innervated by dopamine fibers (Berger et al

1988; Williams and Goldman-Rakic 1993) but relatively

poor in D

2

receptors (Lidow et al 1998), haloperidol

interfered significantly only with spinophilin levels

with-out affecting MAP2 phosphorylation. This suggests that

the cortical dopaminergic system plays an important role

in haloperidol’s ability to affect both the level of

spinophi-lin and phosphorylation of MAP2 and that D

2

receptors

may be particularly involved in regulation of the

phos-phorylation of the latter protein.

Also of interest is that the cortical regions in which

haloperidol treatment affected MAP2 and spinophilin have

all been implicated in the psychopathology of

schizophre-nia (Goldman-Rakic 1999a; Harrison 1999; Weinberger et

al 1994). These regions have also been suggested as

possible sites for the palliative action of antipsychotic

medications (Lidow and Goldman-Rakic 1997; Lidow et

al 1998). It is possible, therefore, that haloperidol-induced

changes in MAP2 phosphorylation and spinophilin levels

may underlie, at least in part, the ability of antipsychotic

medications to improve the quality of life of schizophrenic

patients. It is important to remember, however, that the

results of this study pertain to older (15–25 years of age)

monkeys, and that some of the drug-induced alterations

could be products of an interaction of the treatment with

an aged brain.

The Molecular Biological Findings of This Study

Reflect Morphological Changes in the Cortices of

the Haloperidol-Treated Animals

The molecular biological data collected in this study

correspond well with morphological findings from several

previous studies on the effects of chronic haloperidol

treatment. As mentioned earlier, the degree of MAP2

phosphorylation is inversely proportional to its ability to

stabilize dendritic microtubules, which are essential for the

maintenance of these neuronal processes. Therefore, it is

reasonable to expect that increased MAP2

phosphoryla-tion, which we observed in several cortical regions of

haloperidol-treated animals, would result in some measure

of dendritic collapse in these regions. Indeed, Benes and

her colleagues (Benes et al 1985; Vincent et al 1991)

reported a significant decrease in the number of

small-caliber dendritic processes in the prefrontal cortex of

haloperidol-exposed rats. Furthermore, an electron

micro-scopic analysis (Benes et al 1985) revealed that most of

these processes were dendritic spines, whose necks have a

cytoskeletal structure identical to that of the dendritic shaft

proper (Bloom and Fawcett 1975). These findings are also

compatible with the decrease in levels of the

spine-associated protein spinophilin, which we observed in the

frontal lobe of the drug-treated animals. Additionally, a

decline in the volume of cortical neuropil (which is

composed largely of dendritic processes), together with a

reduced number of microtubules within dendritic shafts

and a lower density of dendritic spines, has been found in

postmortem studies of the frontal cortex of schizophrenic

(9)

patients (Garey et al 1998; Glantz and Lewis 1997;

Selemon et al 1996; Uranova 1988). Our data, along with

those of Benes and her colleagues (Benes et al 1985;

Vincent et al 1991), indicate that antipsychotic treatments

may have contributed to the generation of some of the

aforementioned alterations in the schizophrenic brains.

Another observation of our study is that the levels of

MAP2 in the haloperidol-treated animals, though not

reaching statistical significance, were consistently

ele-vated in the same cortical areas where increases in

phosphorylation of this protein were detected. This is

consistent with reports that an increase in MAP2

phos-phorylation not only reduces its microtubule-stabilizing

capacity but also elevates its resistance to proteolysis

(Alexa et al 1996; Johnson and Foley 1993). Interestingly,

studies in schizophrenic brains have suggested that this

disease may be characterized by an upregulation of

non-phosphorylated MAP2 and downregulation of the total

MAP2 in some cortical areas (Arnold et al 1995; Cotter et

al 1997; Dwork 1997). Our findings demonstrate that

haloperidol can, at least partially, counteract these effects

both by increasing the levels of MAP2 phosphorylation

and by reducing the extent of MAP2 degradation in

cortical cells. Consequently, this may be among the bases

for the therapeutic benefits of antipsychotic drugs.

It has recently been demonstrated that spines are the

main sites of D

1

dopamine receptors in cortical pyramidal

neurons (for a review, see Goldman-Rakic 1999b).

There-fore, any decrease in the number of spines in the cortex

would be expected to affect the expression of D

1

receptors

by these cells. This may explain a decline in the levels of

D

1

receptors detected after long-term neuroleptic

treat-ment in the primate cortex (Lidow et al 1997; Lidow and

Goldman-Rakic 1994) and in the cortex of schizophrenic

patients (Okubo et al 1997; Sedvall and Farde 1996).

Several studies have suggested that haloperidol

treat-ment is accompanied by a shift in the position of synaptic

contacts from dendritic spines to dendritic shafts, without

significantly changing the density of synaptic contacts per

se within the cortex (Benes et al 1985; Klinzova et al 1989,

1990; Meshul et al 1992). In agreement with these

observations, we detected no significant changes in the

levels of the synaptic marker synaptophysin in any of the

cortical areas examined. Our results also support the

earlier report of Nakahara et al (1998), who found no

changes in the levels of synaptophysin messenger RNA in

the prefrontal cortex of rats chronically treated with

haloperidol. On the other hand, Eastwood et al (1997)

detected an increase in the levels of this message in the

frontoparietal cortex of haloperidol-treated rats. It should

be noted, however, that the latter study characterized the

parietal cortex, which was not analyzed by us nor by

Nakahara et al (1998). Finally, the lack of

haloperidol-induced changes in synaptophysin levels observed in our

study suggests that antipsychotic treatment may not be

responsible for the decrease in this protein reported in the

prefrontal and medial temporal cortical regions of

schizo-phrenic patients (Eastwood and Harrison 1995; Glantz and

Lewis 1997; Honer et al 1999; Karson et al 1999;

Perrone-Bizzozero et al 1996).

Possible Cellular Mechanisms of

Haloperidol-Induced Alterations in the Proteins Observed in

This Study

One of the major pharmacologic properties of haloperidol

as an antipsychotic medication is its ability to block D

2

dopaminergic receptors (for a review, see Seeman 1992).

Since D

2

receptors are negatively coupled to adenylyl

cyclase (Hemmings et al 1987), their blockade results in

an increase in the intracellular levels of cyclic adenosine

monophosphate (cAMP; Kaneko et al 1992; Nilsson and

Eriksson 1993), which may persist during long-term

treat-ments despite D

2

receptor upregulation (Lau and Gnegy

1982; Okada et al 1996). Analysis of the literature

sug-gests that elevated intracellular cAMP levels can lead to

all of the alterations detected in the haloperidol-treated

animals in this study. For example, upregulation of cAMP

activates protein kinase A (PKA; Walsh et al 1972), which

is one of the major enzymes responsible for

phosphoryla-tion (Goldenring et al 1985; Sloboda et al 1975; Tsuyama

et al 1986, 1987; Walaas and Nairn 1989). The

involve-ment of the cAMP–PKA pathway in increasing

phosphor-ylation of MAP2 is supported in our study by the fact that

this increase reaches statistical significance only for the

serine residues, the only MAP2 residues known to be

susceptible to PKA-induced MAP2 phosphorylation

(Goldenring et al 1985; Walaas and Nairn 1989).

Elevated levels of cAMP could also lead to a collapse of

dendritic spines and, consequently, to decreases in levels

of spinophilin in the affected neurons. This collapse might

result from destabilization of the microtubule skeleton of

spine necks due to the PKA-induced increase in MAP2

phosphorylation. Alternatively, increased intracellular

lev-els of cAMP may affect the cAMP–PKA–CREB pathway,

which has also been demonstrated to play an important

role in the maintenance of dendritic spines (Martin and

Kandel 1996; Murphy and Segal 1997).

(10)

respect, it is interesting that chronic administration of the

tricyclic antidepressant desipramine, which interferes with

the brain noradrenergic system, also affects

phosphoryla-tion of the cerebral cortical MAP2 on the serine residue

(Miyamoto et al 1997). Of particular significance may be

the ability of haloperidol to act as a noncompetitive

antagonist at

N

-methyl-

D

-asparate (NMDA) receptors

(Coughenour and Cordon 1997; Whittemore et al 1997).

Activation of NMDA receptors has been shown to induce

MAP2 dephosphorylation and increased spine formation

in the cortex (Collin et al 1997; Halpain and Greengard

1990). Haloperidol-induced blockade of NMDA receptors

should therefore result in increased MAP2

phosphoryla-tion and reduced levels of spine-associated proteins, both

of which were observed in our study.

This work was funded by the National Institute of Mental Health Grant No. MH44866 (PSG-R, MSL), the Hoechst Marion Roussel Co. (PSG-R), and the Essel Foundation (MSL).

The authors thank Galina Artamonova, Tatyana A. Trakht, Heather A. Findlay, and Terri A. Beattie for their excellent technical assistance; Dr. Ashiwel S. Undie for his help in designing experiments aimed at analysis of protein phosphorylation; and Dr. Lynn D. Selemon for sharing her extensive expertise in three-dimensional cell counting.

References

Alberts B, Bray D, Lewis J, Raff M, Roberts K, Watson J (1995):

Molecular Biology of the Cell.New York: Garland. Alexa A, Tompa P, Baki A, Vereb G, Friedrich P (1996): Mutual

protection of microtubule-associated protein 2 (MAP2) and cyclic AMP-dependent protein kinase II against mu-calpain.

J Neurosci Res44:438 – 445.

Allen PB, Ouimet CC, Greengard P (1997): Spinophilin, a novel protein phosphatase 1 binding protein localized to dendritic spines.Proc Natl Acad Sci U S A94:9956 –9961.

Anders VN (1978): Ultrastructural features of cerebral synapses in schizophrenic parients.Zh Nevropatol Psikhiatr78:1065–1070. Arnold SE, Lee VM-F, Gur RE, Trojanowski JQ (1995): Abnor-mal expression of two microtubule-associated proteins (MAP2 and MAP5) in specific subfields of the hippocampal formation in schizophrenia. Proc Natl Acad Sci U S A

88:10850 –10854.

Bajjalieh SM, Scheller RH (1995): The biochemistry of neuro-transmitter secretion.J Biol Chem270:1971–1974.

Benes FM, Paskevich PA, Davidson J, Domesik VB (1985): Synaptic rearrangements in medial prefrontal cortex of halo-peridol-treated rats.Brain Res348:15–20.

Berger B, Trottier C, Verney C, Gaspar P, Alvarez C (1988): Regional and laminar distribution of the dopamine and serotonin innervation in the in the macaque cerebral cortex: A radioautoradiographic study.J Comp Neurol273:99 –119. Bloom W, Fawcett DW (1975): The Textbook of Histology.

Philadelphia: Saunders.

Borda T, Genaro AM, Cremaschi G (1999): Haloperidol effect on

intracellular signals system coupled to alpha1-adrenergic recep-tor in rat cerebral frontal cortex.Cell Signal11:293–300. Bourdelais AJ, Deutch AY (1994): The effects of haloperidol and

clozapine on extracellular GABA levels in the prefrontal cortex of the rat: An in vivo microdialysis study. Cereb Cortex4:69 –77.

Brodmann K (1994):Localization in the Cerebral Cortex[Gray LJ, translator]. London: Smith-Gordon.

Cleveland DW, Hoffman PN (1991): Neuronal and glial cy-toskeleton.Curr Opin Neurobiol1:346 –353.

Collin C, Miyaguchi K, Segal M (1997): Dendritic spine density and LTP induction in cultured hippocampal slices.J Neuro-physiol77:1614 –1623.

Cotter D, Kerwin R, Doshi B, Martin CS, Everall IP (1997): Alterations in hippocampal non-phosphorylated MAP2 pro-tein expression in schizophrenia.Brain Res765:238 –246. Coughenour LL, Cordon JJ (1997): Characterization of haloperidol

and trifluperidol as subtype-selective N-methyl-D-aspartate (NMDA) receptor antagonists using [3H]TCP and [3H]ifenprodil

binding in rat brain.J Pharmacol Exp Ther280:584 –592. Csernansky JG (1996):Antipsychotics.New York: Springer Verlag. Dwork AJ (1997): Postmortem studies of the hippocampal

formation in schizophrenia.Schizophr Bull23:385– 402. Eastwood SL, Burnet PW, Harrison PJ (1995): Altered

synapto-physin expression as a marker of synaptic pathology in schizophrenia.Neuroscience66:309 –319.

Eastwood SL, Harrison PJ (1995): Decreased synaptophysin in the medial temporal lobe in schizophrenia demonstrated using immunoautoradiography.Neuroscience69:339 –343. Eastwood SL, Heffernan J, Harrison PJ (1997): Chronic

halo-peridol treatment differentially affects the expression of synaptic and neuronal plasticity-associated genes.Mol Psy-chiatry2:322–329.

Edelmann L, Hanson PI, Chapman ER, Jahn R (1995): Synapto-brevin binding to synaptophysin: A potential mechanism for controlling the exocytotic fusion machine.EMBO J 14:224 – 231.

Garey LJ, Ong WY, Patel TS, Kanani M, Davis A, Mortimer AM, et al (1998): Reduced density on cerebral cortical pyramidal neurons in schizophrenia. J Neurol Neurosurg Psychiatry65:446 – 453.

Glantz LA, Lewis DA (1997): Reduction of synaptophysin immunoreactivity in the prefrontal cortex of subjects with schizophrenia. Regional and diagnostic specificity.Arch Gen Psychiatry54:943–952.

Goldenring JR, Vallano ML, DeLorenzo RJ (1985): Phosphory-lation of microtubule-associated protein 2 at distinct sites by calmodulin-dependent and cyclic-AMP-dependent kinases.

J Neurochem45:900 –905.

Goldman-Rakic PS (1999a): The physiological approach: Func-tional architecture of working memory and disordered cogni-tion in schizophrenia.Biol Psychiatry26:650 – 661. Goldman-Rakic PS (1999b): The “psychic” neuron of the

cere-bral cortex.Ann N Y Acad Sci868:13–26.

Halpain S, Greengard P (1990): Activation of NMDA receptors induces rapid dephosphorylation of the cytoskeletal protein MAP2.Neuron5:237–246.

(11)

critical review of the data and their interpretation. Brain

122:593– 624.

Hemmings HC, Walaas SI, Ouimet CC, Greengard P (1987): Dopamine receptors: Regulation of protein phosphorylation. In: Crees I, Fraser CM, editors.Dopamine Receptors.New York: Liss, 115–152.

Honer WG, Falkai P, Chen C, Arango V, Mann JJ, Dwork AJ (1999): Synaptic and plasticity-associated proteins in anterior frontal cortex in severe mental illness. Neuroscience 91: 1247–1255.

Honer WG, Kaufmann CA, Davies P (1992): Characterization of a synaptic antigen of interest in neuropsychiatric illness.Biol Psychiatry31:147–158.

Hutz RJ, Dierschke DJ, Wolf RC (1985): Seasonal effects on ovarian folliculogenesis in rhesus monkeys. Biol Reprod

33:653– 659.

Johnson GV, Foley VG (1993): Calpain-mediated proteolysis pf microtubule-associated protein 2 (MAP2) is inhibited by phosphorylation by cAMP-dependent protein kinase but not by CA21/calmoduline-dependent protein kinase II.J Neuro-sci Res34:642– 647.

Johnson GVW, Jope RS (1992): The role of microtubule-associated protein 2 (MAP2) in neuronal growth, plasticity, and degeneration.J Neurosci Res33:505–512.

Kaneko M, Sato K, Hirikoshi R, Yaginuma M, Yaginuma N, Shiragata M, Kumashiro H (1992): Effects of haloperidol on cyclic AMP and inositol triphosphate in rat striatumin vivo. Prostaglandins Leukot Essent Fatty Acids46:53–57. Karson CN, Mark RE, Schluterman KO, Sturner WQ, Sheng JG,

Griffinj WS (1999): Alterations in synaptic proteins and their encoding mRNAs in prefrontal cortex in schizophrenia: A possible neurochemical basis for “hypofrontality”.Mol Psy-chiatry4:39 – 45.

Kebabian JW, Greengard P (1971): Dopamine-sensitive adenyl cyclase: Possible role in synaptic transmission.Science174: 1346 –1349.

Keith CH (1990): Neurite elongation is blocked if microtubules polymerization is inhibited in PC12 cells. Cell Motil Cy-toskeleton17:95–105.

Kelley JJ, Gao XM, Tamminga CA, Roberts RC (1997): The effect of chronic haloperidol treatment on dendritic spines in the rat striatum.Exp Neurol146:471– 478.

Klinzova AJ, Haselhorst U, Uranova NA, Schenk H, Isomin VV (1989): The effect of haloperidol on synaptic plasticity in rat medial prefrontal cortex.J Hirnforsch30:51–57.

Klinzova AJ, Uranova NA, Haselhorst U, Schenk H (1990): Synaptic plasticity in rat medial prefrontal cortex under haloperidol treatment produced behavioral sensitivity.J Hirn-forsch2:173–179.

Lau Y-S, Gnegy ME (1982): Chronic haloperidol treatment increased calcium-dependent phosphorylation in rat striatum.

Life Sci30:21–28.

Lidow MS, Elthworth JD, Goldman-Rakic PS (1997): Down-regulation of the D1 and D5 dopamine receptors in the primate prefrontal cortex by chronic treatment with antipsy-chotic drugs.J Pharmacol Exp Ther281:597– 603.

Lidow MS, Goldman-Rakic PS (1994): A common action of clozapine, haloperidol and remoxipride on D1-and D2-dopa-mine receptors in the primate cerebral cortex.Proc Natl Acad Sci U S A91:4353– 4356.

Lidow MS, Goldman-Rakic PS (1997): Differential regulation of D2 and D4 dopamine receptor mRNAs in the primate cerebral cortex vs. neostriatum: Effects of chronic treatment with typical and atypical antipsychotic drugs. J Pharmacol Exp Ther283:939 –946.

Lidow MS, Goldman-Rakic PS, Rakic P, Innis RI (1989): Dopamine D2 receptors in the cerebral cortex: Distribution and pharmacological characterization with [3H]raclopride.

Proc Natl Acad Sci U S A86:6412– 6416.

Lidow MS, Williams G, Goldman-Rakic PS (1998): A case for cerebral cortex as a common site of action by antipsychotic medications.Trends Pharmacol Sci19:136 –140.

Lipton SA, Kater SB (1989): Neurotransmitter regulation of neuronal outgrowth, plasticity and survival.Trends Neurosci

7:265–270.

Maccioni RB, Cambiazo V (1995): Role of microtubule-associ-ated proteins in the control of microtubule assembly.Physiol Rev75:835– 864.

Martin KC, Kandel ER (1996): Cell adhesion molecules, CREB, and the formation of new synaptic connections. Neuron

17:567–570.

Masliah E, Terry RD, Alford M, DeTersa R (1990): Quantitative immunohistochemistry of synaptophysin in human neocortex: An alternative method to estimate density of presynaptic termi-nals in paraffin sections.J Histochem Cytochem38:837– 844. Meshul CK, Janowsky A, Casey DE, Stallbaumer RK, Taylor B

(1992): Effects of haloperidol and clozapine on the density of “perforated” synapses in caudate, nucleus accumbens, and medial prefrontal cortex.Psychopharmacology106:45–52. Miyamoto S, Asakura M, Sasuga Y, Osada K, Bodaiji N,

Imafuku J, Aoba A (1997): Effects of long-term treatment with desipramine on microtubule proteins in rat cerebral cortex.Eur J Pharmacol333:279 –287.

Murphy DD, Segal M (1997): Morphological plasticity of dendritic spines in central neurons is mediated by activation of cAMP resdponse element binding protein.Proc Natl Acad Sci U S A94:1482–1487.

Nakahara T, Nakamura K, Tsutsumi T, Hashimoto K, Hondo H, Hisatomi S, et al (1998): Effects of chronic haloperidol treatment on synaptic protein mRNAs in the rat brain. Mol Brain Res61:238 –242.

Nalepa I (1993): The effect of chlorpromazine and haloperidol on second messenger system related to adrenergic receptors.

Pol J Pharmacol45:399 – 412.

Nieuwenhuys R (1985):Chemoarchitecture of the Brain. New York: Springer Verlag.

Nilsson CL, Eriksson E (1993): Haloperidol increases prolactin release and cyclic AMP formationin vitro:Inverse agonism at dopamine D2 receptors?J Neural Transm92:213–220. Okada F, Ito A, Horikawa T, Tokumitsu Y, Nomura Y (1996):

Long-term neuroleptic treatments counteract dopamine D2 agonist inhibition of adenylate cyclase but do not affect pertussis toxin ADP-rybosylation in the rat brain.Neurochem Int28:161–168.

Okubo Y, Suhara T, Suzuki K, Kabayashi K, Inoue O, Terasaki O, et al (1997): Decreased prefrontal dopamine D1 receptors in schizophrenia revealed by PET.Nature385:634 – 636. Perrone-Bizzozero NI, Sower AC, Bird ED, Benowitz LI, Ivins

(12)

GAP-43 are selectively increased in association cortices in schizophrenia.Proc Natl Acad Sci U S A93:14182–14187.

Physicians’ Desk Reference,53rd ed (1999): Oradell, NJ: Med-ical Economics.

Roth BL, Berry SA, Kroeze WK, Willins DL, Kristiansen K (1998): Serotonin 5HT2A receptors: Molecular biology and mechanisms of regulation.Crit Rev Neurobiol12:319 –338. Sasaki T, Kennedy JL, Nobrega JN (1997): Localized changes in

GABA receptor-gated chloride channel in rat brain after long-term haloperidol:Relation to vacuous chewing move-ments.Synapse25:73–79.

Sedvall G, Farde L (1996): Dopamine receptor in schizophrenia.

Lancet347:264.

Seeman P (1988): Tardive dyskinesia, dopamine receptors, and neuroleptic damage to cell membranes. J Clin Psychophar-macol8(suppl):3S–9S.

Seeman P (1990): Atypical neuroleptics: Role of multiple recep-tors, endogenous dopamine, and receptor linkage.Acta Psy-chiatr Scand358(suppl):14 –20.

Seeman P (1992): Dopamine receptor sequences. Therapeutic levels of neuroleptics accupy D2 receptors, clozapine occu-pies D4.Neuropsychopharmacology7:261–284.

Selemon LD, Lidow MS, Goldman-Rakic PS (1999): Increased volume and glial density in primate prefrontal cortex associ-ated with chronic antipsychotic drug exposure. Biol Psychi-atry46:161–172.

Selemon LD, Rajkowska G, Goldman-Rakic PS (1996): Abnor-mally high neuronal density in the schizophrenic cortex.Arch Gen Psychiatry52:805– 818.

Shea TB, Beermann ML (1994): Respective roles of neurofila-ments, microtubules, MAP1B and tau in neurite outgrowth and stabilization.Mol Cell Biol5:863– 875.

Sloboda RD, Rudolph SA, Rosenbaum JL, Greengard P (1975): Cyclic AMP-dependent endogenous phosphorylation of a microtubule-associated protein. Proc Natl Acad Sci U S A

72:177–181.

Smith FD, Oxford GS, Milgram SL (1999): Association of D2 dopamine receptor third cytoplasmic loop with spinophilin, a protein phosphatase-1-interacting protein. J Biol Chem274: 19894 –19900.

Tsuyama S, Bramblet GT, Huang KP, Flavin M (1986): Calcium/ phospholipid-dependent kinase recognized sites in microtu-bule-associated protein 2 which are phosphorylated in living brain and are not accesible to other kinases. J Biol Chem

261:4110 – 4116.

Tsuyama S, Terayama Y, Matsuyama S (1987): Numerous phosphates of microtubule-associated protein in living rat brain.J Biol Chem262:10886 –10892.

Uranova NA (1988): Structural changes in the neuropil of the frontal cortex in schizophrenia.Zh Nevropatol Psikhiatr88:52–58. Uranova NA, Casanova MF, DeVaughn NM, Orlovskaya DD,

Denisov DV (1996): Ultrastructural alterations of synaptic contactes and astrocytes in postmortem caudate nucleus of schizophrenic patients.Schizophr Res22:81– 83.

Uranova NA, Orlovskaya DD, Apel K, Klintsova AJ, Haselhorst U, Schenk H (1991): Morphometric study of synaptic patterns in the rat caudate nucleus and hippocampus under haloperidol treatment.Synapse7:253–259.

Vincent SL, Adamec E, Sorensen I, Benes FM (1994): The effects of chronic haloperidol administration on GABA-immunoreactive axon terminals in rat medial prefrontal cor-tex.Synapse17:26 –35.

Vincent SL, McSparren J, Wang RY, Benes FM (1991): Evi-dence for ultrastructural changes in cortical axodendritic synapses following long-term treatment with haloperidol or clozapine.Neuropsychopharmacology5:147–155.

Walaas SI, Nairn AC (1989): Multisite phosphorylation of microtubule-associated protein 2 (MAP2) in rat brain: Peptide mapping distinguishes between cyclic AMP-, calcium/cal-modulin-, and calcium/phospholipid-regulated phosphoryla-tion mechanisms.J Mol Neurosci1:117–127.

Walker E (1940): A cytoarchitectural study of the prefrontal area of the macaque monkey.J Comp Neurol73:59 – 86. Walker ML, Gordon TP, Wilson ME (1983): Menstrual cycle

characteristics of seasonally breeding rhesus monkeys. Biol Reprod29:841– 848.

Walsh DA, Brostrom CO, Brostrom MA, Chen L, Corbin JD, Reimann E, et al (1972): Cyclic AMP-dependent protein kinase from skeletal muscle and liver.Adv Cyclic Nucleotide Res1:33– 45.

Weinberger DR, Aloia MS, Goldberg TE, Berman KF (1994): The frontal lobes and schizophrenia.J Neuropsychiatry Clin Neurosci6:419 – 427.

Whittemore ER, Ilyin VI, Woodward RM (1997): Antagonism of N-methyl-D-aspartate receptors by s site ligands: Potency, subtype-selectivity and mechanisms of inhibition.J Pharma-col Exp Ther282:326 –338.

Wiche G, Oberkanins C, Himmler A (1991): Molecular structure and function of microtubule-associated proteins.Int Rev Cytol

124:217–273.

Williams RW, Rakic P (1988a): Elimination of neurons from the rhesus monkey’s lateral geniculate nucleus during develop-ment.J Comp Neurol272:424 – 436.

Williams RW, Rakic P (1988b): Three-dimentional counting: An accurate and direct method to estimate numbers of cells in sectioned material.J Comp Neurol278:344 –352.

Williams SM, Goldman-Rakic PS (1993): Characterization of the dopaminergic innervation of the primate frontal cortex using a dopamine-specific antibody.Cereb Cortex3:199 – 222.

Wilson JR, Ludowyke RS, Biden TJ (1998): Nutrient stimulation results in a rapid Ca12-dependent threonine phosphorylation

of myosin heavy chain in rat pancreatic islets and RINm5F cells.J Biol Chem28:22729 –22737.

Yamada KM, Spooner BS, Wessells NK (1970): Axon growth: Role of microfilaments and microtubules.Proc Natl Acad Sci U S A66:1206 –1212.

Yan Z, Hsieh-Wilson L, Feng J, Komizawa K, Allen PB, Fienberg AA, et al (1999): Protein phosphatase 1 modulation of neostriatal AMPA channels: Regulation by DARPP-32 and spinophilin.Nat Neurosci2:13–17.

Gambar

Figure 1. Diagrams of the lateral, medial, and ventral surfaces ofthe rhesus monkey cerebral hemisphere on which the positions ofthe regions examined in this study have been indicated
Figure 2. Representative examples of the images of the Westernblots and slot blots generated in this study.images generated by the spinophilin immunolabeling of slot blotsamples from tissue homogenates of the right dorsolateral pre-frontal cortex from four
Figure 3. Film images of Western blots of a set of MAP2immunoprecipitates from the right hemisphere dorsolateral pre-frontal cortex of control animals (1–4) and haloperidol-treated(5–8) animals
Figure 4. Histograms showing differences in the levels ofMAP2, spinophilin, and synaptophysin between the control andof the mean levels of these proteins in control samples
+2

Referensi

Dokumen terkait

DILEMA MASYARAKAT SUMEDANG TERHADAP KEHADIRAN GERAKAN DI/TII TAHUN 1950-1962.. Universitas Pendidikan Indonesia | repository.upi.edu | perpustakaan.upi.edu

PROGRAM ADIWIYATA adalah program untuk mewujudkan sekolah yang peduli dan berbudaya lingkungan.. SEKOLAH ADIWIYATA : adalah sekolah peduli dan

mengalami peningkatan kandungan asam laktat seiring dengan peningkatan lama inkubasi, hingga lama inkubasi 10 hari, dan selanjutnya mengalami stagnasi sampai lama inkubasi

Daftar Peserta Seleksi Profile Assessment dan Wawancara Calon Hakim Ad Hoc Pengadilan Tindak Pidana Korupsi Tahap IV sebagai berikut

[r]

Buku/Dokumen Manual SPMI adalah dokumen berisi petunjuk teknis tentang cara, langkah, atau prosedur Penetapan, Pelaksanaan, Evaluasi pelaksanaan,

Standar Pendidikan Tinggi yang Ditetapkan oleh Perguruan.. Tinggi adalah sejumlah standar pada perguruan

Dugaan bahwa kemampuan pemecahan masalah, kemampuan berpikir dan sikap mahasiswa dipengaruhi oleh faktor pembelajaran dan kemampuan awal cukup beralasan mengingat