You are on page 1of 23

Immediate-Early

Genes
as Activity Markers in the CNS
George

I. Discovery

S. Robertson

of the Proto-Oncogenes

Genetic analysis of viruses capable of producing tumors in mice


led to the discovery of cancer-causing genes termed oncogenes.
The v-fos oncogene is responsible for the ability of the FBJ-MSV
virus to produce bone tumors (Finkel et al., 1966; Curran and Teich,
1982). Shortly after identification of v-fos, it became clear that this
oncogene had a normal cellular counterpart (Curran et al., 1984).
The normal cellular sequences from which the viral oncogene
(V-$X) was derived is referred to as thefos proto-oncogene or c-fos.
The protein product of c-fos is a 55-kDa protein (Fos) that plays an
important role in the signal transduction events mediating cell
growth and division (Morgan and Curran, 1991). Proto-oncogenes
such as c-fos contain negative regulatory elements that prevent
overexpression (Sassone-Corsi et al., 1988; Gius et al., 1990). However, these expression-limiting
elements are not present within
v-fos, enabling the FBJ-MSV virus to produce osteosarcomas (bone
tumors). Overexpression of oncogene products in virally infected
cells leads to tumor formation because the signal transduction
pathways specifying growth and division become overstimulated
(Carbone and Levine, 1990).
The c-fus and c-jun proto-oncogenes were identified as genes
whose rapid but transient transcription was activated by exposure of cells to serum or growth factors that initiate the cell cycle
(Greenberg and Ziff, 1984; Lamph et al., 1988). A number of related
proto-oncogenes were discovered shortly thereafter, using probes

From
Neuromethods,
vol
Eds A A Boulton,
G B Baker,

33

Cell

Neurobiology

and A N Bateson

231

Techmques

0 Humana

Press lnc

232

Robertson

based upon sequences found within these genes to screen cDNA


libraries constructed
from serum-stimulated
cells. These included
f&B and thefts-related
antigens (fva-1 andfia-2)
as well as jun-B
(Cohen and Curran, 1988; Lau and Nathans,
1987; Ryder et al.,
1988; Zerial et al., 1989; Mshina
et al., 1990). Although
jUM-D
expression
was not markedly
elevated
by growth
factors or
serum, its high constitutive
expression in 3T3 cells permitted
isolation of this third member
of the jun family (Hirai et al., 1989;
Ryder et al., 1989). The zinc finger-containing
gene NGFI-A,
also
known as zip68, krox-24 and egr-1, was identified
as a gene that is
rapidly
activated
by growth
factors or serum in 3T3 cells
(Milbrandt,
1987).

2, Regulation
2.7.

of c-fos Expression

c-fos is an Immediate-Early

Gene

In most cells, basal expression


of c-fos mRNA and protein
is
low (Morgan et al., 1987; Sagar et al., 1988; Smeyne et al., 1992). In
such cell types, extracellular
signals are required
to elevate
expression
of this proto-oncogene.
It is now well established
that
c-fos expression
in the central nervous system (CNS) can be triggered by a broad host of physiological
and pharmacological
treatments that increase neuronal
activity (Morgan
and Curran, 1991;
Hughes and Dragunow,
1995). This has led to the wide spread
use of c-fos as a metabolic
marker for mapping
functional
pathways in the CNS. These studies have shown that the time course
for induction
of c-fos expression
is similar
in most cases. At the
transcriptional
level, activation
usually takes place within several
minutes and lasts for approx 20 min with peak increases in mRNA
occuring 30-45 min after stimulation
(Muller
et al., 1984). After
this time, mRNA levels rapidly decrease with a half-life of approx
12 min. Synthesis
of Fos follows mRNA
expression
with peak
increases detectable
approx l-2 h after the onset of stimulation,
thereafter Fos levels rapidly decline to basal levels by 6-8 h (Muller
et al., 1984; Curran
et al., 1984; Sonnenberg
et al., 1989). The
induction
of c-J?OStranscription
is not dependent
on the synthesis
of new proteins
and readily occurs m the prescence of protein
synthesis inhibitors
(Lau and Nathans, 1987; Curran and Morgan,
1986). This indicates
that the protems required
for c-fos expression are present in unstimulated
cells and that their activation
is
mediated
by posttranslational
processes such as phosphorylation.

Immediate-Early

Genes as Activity

Markers

233

Since transcriptional induction in the presence of protein synthesis inhrbitors is characteristic of viral immediate-early genes, c-fos
and other rapidly induced genes, are commonly referred to as cellular immediate-early genes (Lau and Nathans, 1987; Curran and
Morgan, 1987).
2.2. The Calcium Response Element
The first demonstration that neurotransmitters
can activate
immediate-early gene (IEG) expression came from studies showing that depolarization of rat PC 12 pheochromocytoma cells by
exposure to nicotine produces a rapid elevation of c-f& expression (Greenberg et al., 1986). Elegant studies performed by
Greenberg and colleagues have yielded significant insights into
the signal transduction events that mediate c-fos activation by
depolarizing neurotransmitters. A key step in this process is the
influx of Ca2+ions through specialized channels embedded in the
plasma membrane. In neurons, calcium entry may occur by way
of at least two types of Ca2+channels: voltage sensitive calcium
channels (VSCCs) and N-methyl N-aspartate (NMDA) receptors.
In the case of VSCCs, channel opening is triggered by membrane
depolarization. In contrast, NMDA receptors are ligand-gated ion
channels that require both occupation by ligand and membrane
depolarization to open. The subsequent rise in intracellular Ca2+
induces c-fos transcription (Morgan and Curran, 1986; Curran and
Morgan, 1986). A Ca*+ response element (CaRE) locateld 60 nucleotides from the 5 initiation site for c-fos mRNA synthesis plays
an important role in mediating the c-fos response to VSCC activation (Sheng et al., 1990). The CaRE (-TGACGTTT-)
is similar
in sequence to a consensus CAMP response element (CRE)
(-TGACGTCA-) located within the regulatory regions of a variety
of genes that become activated when cells are exposed to agents
such as forskolin that activate adenylate cyclase and stimulate the
production of CAMP (Montminy et al., 1986). Placement of the
c-fos CaRE/CRE into the promotor of genes that fail to respond to
forskolin or VSCC activators endows the ability to respond to these
agents (Sheng et al., 1990; Sheng et al., 1991). Constitutively
bound to the -60 CaRE is the calcium response element binding
protein (CREB) that is converted into a positive transcriptional
factor by phosphorylation
at a critical regulatory site, serine 133
(Sheng et al., 1990). A crucial role for serine 133 phosphorylation in the activation of CREBs transcriptional
stimulating

234

Robertson

activity is indicated by the loss of this function when serine 133


is mutated to alanine (Sheng et al., 1991; Gonzales and
Montminy, 1989; Schwaninger et al., 1993). Serine 133 of CREB
may be phosphorylated
by calcium/calmodulin-dependent
protein kineses (CaM kinases) as well as CAMP-dependent
kinase (PKA); both of these classes of kinases are thought to play
a role in mediating the induction of c-fos expression by VSCC
activators (Ghosh and Greenberg, 1995).
2.3. The Serum Response Element
A second key regulatory element in the c-fos promotor that
confers second messenger inducibility of this IEG is the serumresponse element (SIRE). The SRE was originally described as a
protein-binding
site required for the induction of c-fos expression
by serum and growth factors (Treisman, 1992). The SRE, together
with flanking DNA sequences, binds an assembly of multiprotein
complexes that include the serum-response factor, Elk-l, and several other transcriptional regulating factors (Shaw et al., 1989;
Hipskind et al., 1991; Hill et al., 1993). At present, the precise
mechanism responsible for activation of c-fos transcription via the
SRE is unclear, but likely involves a Ras-dependent mechanism
that culminates in phosphorylation
of Elk-l by microtubuleassociated protein (MAP-11 (Marais et al., 1993). The development
of techniques enabling transfection of primary neurons has
assisted analysis of the relative roles of the CaRE and SRE in
NMDA receptor-mediated
signaling. In hippocampal neurons,
NMDA receptor activation does not trigger significant amounts
of CaRE-dependent transcription (Bading et al., 1993). Nevertheless, Ca*+ influx through the NMDA receptor does result in the
induction of C--OSas well as several other IEGs in hippocampal
neurons indicating mediation by a non-CaRE element such as
the SRE. Consistent with this proposal, transfection studies performed on cultured hippocampal
neurons have shown that
NMDA receptor activation of C-$X requires phosphorylation
of
the SRE. In contrast to NMDA-mediated
c-f0.s expression, activation of VSCCs by elevation of extracellular KC1 concentrations
promotes c-fos transcription by phosphorylation
of CREB and
occurs in the absence of the SRE (Bading et al., 1993). These findings suggest that Ca*+ influx through VSCCs and NMDA receptors leads to the activation of c-fos expression via distinct
signalling pathways.

Immedate-Early

Genes as Actwity

Markers

23.5

2.4. Regulatory Elements in the c-fos Promotor


Operate in an Interdependent
Fashion
The previously described studies that were performed in cultured cells suggest that the regulation of c-fos expression is mediated by individual response elements (CaRE and SRE) that act
independently in response to extracellular stimuli. However, in
the intact organism, it is unlikely that the independent actions of
individual regulatory elements can account for the broad range
of stimuli which can induce c-fos expression. This issue was
recently addressed using a Fos-1acZ transgenic mouse in which
expression of the bacterial J3-galactosidase gene is driven by the
c-fos promoter. The Fos-1acZ transgene directs inducible expression of a fusion protein consisting of 315 N-terminus amino acids
from c-fos and 1015 C-terminus amino acids from /3-galactosidase
(Schilling et al., 1991; Smeyne et al., 1992). The Fos-1acZ fusion
protein retains the nuclear localization signal for Fos, and
P-galactosidase is exclusively revealed in nuclei. Employing the
histochemical detection of P-galactosidase activity encoded by this
gene, Smeyne et al. (1992) have shown that the Fos-1acZ construct
recapitulates both tissue- and stimulus-specific
regulation of
c-fos expression in vivo. In order to determine the role of the CaRE
and SRE sites in controlling c-fos expression in the intact organism, transgenic animals have been created in which these
regulatory elements in the Fos-1acZ construct were rendered nonfunctional by the introduction of clustered point mutations. Consistent with transient transfection
experiments in cultured
hippocampal neurons, mutation of the CaRE abolished Fos-1acZ
induction in primary neuronal cultures by KC1 (Robertson et al.,
199513).However, mutation of the SRE also eliminated KCl-induced
c-fos expression suggesting that multiple elements are necessary
and none sufficient for the complete activation of the gene by KCl.
A similar finding occurred in vivo. For instance, the induction of
c-fos in response to kainate-induced seizures is thought to involve
several transduction pathways, particularly Ca*+ influx through
VSCCs. However, mutation of either the CaRE or SRE completely
abolished karinate-induced c-fos expression in the majority of neurons (Robertson et al., 1995b). Interestingly, the excitatory effects of
both KC1 and kainate on neuronal c-fos expression were also lost
after selective mutation of the &-inducible element (SIE). The SIE
is a regulatory element found within the c-fos promoter that is

236

Robertson

thought to confer inducibility


of this gene to platelet derived
growth factor (PDGF) (Wagner et al., 1990). Consequently, the
CaRE, SRE, and SIE were required in combination for induction
of c-fos expression in many neurons. These findings suggest that,
at least within the context of the c-fos promoter, physiological signals in the CNS are not transduced in a linear fashion, resulting in
activation of a single response element but rather by interdependent networks of transcriptional regulating factors that require
multiple regulatory elements to operate properly (Robertson et
al., 1995b).

3. IEGs as Activity

Markers in the CNS

3. I. Overview
Although basal levels of c-fos mRNA and protein are low in the
CNS, neuronal expression of this IEG is rapidly but transiently
elevated by a broad array of extracellular stimuli. Since a complete description of the physiological and pharmacological treatments that induced c-fos expression is beyond the scope of the
present review, I will focus upon those examples which are of
relevance to neuropsychopharmacology.
As indicated previously,
the c-fos promoter contains regulatory elements that are activated
by the second messengers AMP and Ca2+. Since generation of
these second messengers is linked to stimulation of the extracellular receptors for a broad range of neurotransmitters, detection
of c-fos mRNA and protein has proven to be a quick, inexpensive,
and reliable method for the identification of putative neuronal
targets for various classes of neuropharmacological
agents.
Moreover, double labeling Fos-positive neurons with classic
neurochemical markers or retrograde tracers has permitted characterization of the phenotypic and connection character of neurons that express this IEG. Lastly, inhibition of c-fos expression
using antisense DNA technology has revealed some of the physiological targets for this transcriptional regulating factor.
3.2. EC Induction in the Forebrain
by Treatments that Induce Seizures
Administration
of pentylenetetrazole (PTZ) to rodents results
in the rapid onset of seizures and convulsions that last for approx
30 min. Within minutes of PTZ-induced seizures, a synchronous

Immediate-Early

Genes as Activity

Markers

237

wave of IEG expression occurs that is localized to those brain


regions which display seizure activity such as the hippocampus,
cortex and limbic system (Morgan et al., 1987; Saffen et al., 1988;
Sonnenberg et al., 1989a,b; Le Galle La Salle and Naquet, 1990).
PTZ-induced seizures result in the elevation of c-fos, c-jun, jun-B,
jun-D, and NGF-A in all of these brain regions. In addition to PTZ,
elevated neuronal IEG expression has been reported after the
induction of seizures by kindling (Dragunow and Robertson,
1987), electroconvulsive
shocks (Hope et al., 1994a), audogenic
stimulation (Le Galle La Salle and Naquet, 1990), kainic acid
(Popovici et al., 1988), bicuculline (Gass et al., 1992a), or pilocarpine
(Barone et al., 1993). Seizure-induced IEG expression is paralleled by
an elevation of AP-l-like DNA binding that persists for up to 8 h
after the initiation of seizures (Sonnenberg et al., 1989a,b). Western
blotting performed with antisera that recognizes all known members of the Fos family indicates that Fos production peaks 1-2 h
after the onset of seizures and returns to basal levels by 6-8 h.
This indicates that Fos does not participate in Al?-1 complexes
detected at these later time points. In contrast, several lower
molecular weight proteins (35 kDa and 46 kDa) detected with this
antisera, termed Fos-related antigens (Fras), display prolonged
induction kinetics with respect to Fos. Subsequent studies have
shown that the 46-kDa Fra is actually FosB whereas the 35-kDa
Fra corresponds to a truncated version of FosB known AFosB
(Hope et al., 1994b). Using FosB and AFosB-selective antibodies,
it has been demonstrated that after seizure onset FosB expression peaks by 2-4, whereas AFosB levels are maximal at 4 h and
remain elevated for at least 8 h. Hence, seizures stimulate the formation of dynamic AP-1 complexes whose composition changes over
time. Initially, seizure-induced Al?-1 complexes consist primarily
of Fos/ Jun dimers, but are replaced by FosB/ Jun and AFosB/Jun
dimers at later time points.
MK-801 is a noncompetitive
antagonist of the N-methylD-aspartate (NMDA) subclass of excitatory aminoacid receptors.
The distribution of NMDA receptors in the brain closely matches
the distribution of neurons that express Fos-like immunoreactivity after administration of PTZ suggesting that activation of these
receptors mediates c-fos induction by seizure activity (Morgan et
al., 1987). Consistent with this proposal, glutamate receptor agonists increase c-fos expression in the brain whereas MK-801 reduces
IEG activation after kindling-induced
seizures (Sonnenberg et al.,

238

Robertson

1989b; Page and Everitt, 1993; Sugimoto et al., 1993). The failure
of MK-801 to block IEG expression produced by electroshock (Cole
et al., 1990a) and lindane-induced seizures (Vendrell et al., 1992)
suggests that the activation of non-NMDA
receptors and/or
VSCCs may also play an important role in this process. In addition to seizures, MK-801 has been reported to reduce IEG expression produced by more physiologically
relevant stimuli. For
instance, NMDA receptor blockade prevents light-induced c-jiis
and NGFI-A mRNA expression m the retina (Gudehithlu
et
a1.,1993). MK-801 also abolishes the high constitutive expression
of zif268 (NGFI-A) in the cortex indicating that expression of this
IEG is driven by natural synaptic activity (Worley et al., 1991).
3.3. IEG Induction in the Spinal Cord
by Nonnoxious and Painful Stimulation
Physiological stimulation of rat primary sensory neurons by hair
brushing or gentle joint manipulation promotes a modest elevation of Fos-like immunoreactivity
in nuclei of postsynaptic neurons of the dorsal horn (Hunt et al., 1987). These increases occur
mainly in layers II-IV which are innervated by low-threshold
AGcutaneous afferents. In contrast, painful chemical or heat stimulation of the hind paws markedly increases Fos-like immunoreactivity in layers I and II of the dorsal horn, which receive excitatory
input from nociceptive afferent terminals (Hunt et al., 1987).
Noxious peripheral stimulation also results in the appearance of
Fos expression in thalamic regions known to process painful
stiumulation (Bullitt, 1989). Consistent with the notion that these
increases are related to the activation of pain pathways, administration of morphine substantially reduces the induction of Fos-like
immunoreactivity
in superficial layers of the dorsal horn by
noxious stimulation of the hind paw (Tolle et al., 1990).
3.4. IEG Induction

in the Striatum

by Dopaminergic

Drugs

Basal c-fos expression in the striatum in very low but is rapidly


elevated by systemic administration of cocaine and d-amphetamine,
stimulants that greatly enhance extracellular concentrations of
dopamine (Robertson et al., 1989b; Graybiel et al., 1990). In contrast, levodopa and directly acting Dl-like receptor agonists such
as SKF 38393 and CY 208-243 only weakly increase Fos-like
immunoreactivity
in the intact striatum (Robertson et al., 1989a,b;

Immediate-Early

Genes as Activity

Markers

239

Robertson et al., 1992). Destruction of the nigrostriatal pathway


with 6-OHDA, however, endows levodopa and Dl-like agonists
with the ability to dramatically enhance c-fos expression in the
denervated striatum (Robertson et al., 1989a,b; Paul et al., 1992;
Morelli et al., 1993). These increases are completely blocked by
the selective Dl-like receptor antagonist SCH 23390 indicating that
activation of Dl-like receptors plays an important role in levodopaand Dl-like agonist-induced c-fos expression (Robertson et al.,
1989b, Morelli et al., 1993). The failure of levodopa and Dl-like
agonists to increase Fos-like immunoreactivity
in the striatum of
intact animals suggests that the development of denervation-induced
supersensitivity
is responsible for the large increases in c-fos
expression produced by these compounds in the 6-OHDA-denervated
striatum. This is consistent with the proposal that postsynaptic
changes in the striatum may contribute to the development of
levodopa-induced
dyskinesias in Parkinsons disease (Chase et
al., 1993; Obseo et al., 1994).
D2-like receptor antagonists such as haloperidol and raclopride
increase c-fos expression in the intact striatum (Dragunow et al.,
1990; Deutch et al., 1992; Robertson and Fibiger, 1992). The distribution of increased Fos-like immunoreactivity
produced by these
neuroleptics very closely matches the distribution of striatal D2
receptors (Boyson et al., 1986; Robertson and Fibiger, 1992). The
close topographical
relationship between neuroleptic-induced
Fos-like immunoreactivity
and D2 receptors suggests that haloperidol and raclopride increase Fos-like immunoreactivity
in strratal neurons that express D2 receptors, that is, in striatopallidal
neurons (Gerfen and Young, 1988; Gerfen et al., 1990). The fact
that enkephalin is utilized principally by striatopallidal neurons
as a neurotransmitter,
enabled us to preferentially label striatopalhdal with an oligonucleotide probe complementary to mRNA
encodmg enkephalin. Thus, by combining Fos-like immunohistochemistry with the detection of proenkephalin mRNA by tn situ
hybridization histochemistry, we were able to demonstrate that
D2 antagonists elevate Fos-like immunoreactivity in striatopallidal
neurons (Robertson et al., 1992). Furthermore, neuroleptic-induced
Fos-like immunoreactivity
was seldom found in striatonigral neurons retrogradely labeled with fluoro-gold from the substantia
nigra pars reticulate (Robertson et al., 1992). These findings indicate that D2-like receptor antagonists elevate c-fos expression primarily in striatopallidal neurons. Moreover, they are consistent

240

Robertson

with neurochemical and neurophysiological studies showing that


dopamine inhibits striatopallidal activity (Pan et al., 1985; Gerfen
et al, 1990. Carlson et al., 1990).
Using retrograde tracing techniques, we have demonstrated that
the Dl-like receptor agonist SKF 38393 elevates Fos-like immunoreactivity in striatonigral neurons (Robertson et al., 1990). This
result is consistent with 2-deoxyglucose uptake studies showing
that Dl-like receptor agonists activate striatonigral neurons ipsilateral to the 6-hydroxydopamine-lesioned
substantia nigra
(Trugman and Wooten, 1987). In order to determine whether SKF
38393-induced Fos-like immunoreactivity
was also present in
striatopallidal neurons, we combined Fos-like immunohistochemistry with the detection of proenkephalin
mRNA by in situ
hybridization histochemistry. Dl-like receptor-activated Fos-like
immunoreactivity
was infrequently found in striatopallidal neurons identified with the proenkephalin
oligonucleotide
probe
indicating that it is preferentially localized in striatonigral neurons (Robertson et al., 1992). This finding is in agreement with
autoradiographic
and in sztu hybridization studies which report
that Dl receptors are expressed predominantly
by striatonigral
neurons (Harrison et al., 1990; Gerfen et al., 1990; Le Moine et al.,
1991). Taken together, these results suggest Dl-like agonist-induced
c-fos expression may be utilized as a measure of striatonigral
activation.
In contrast to levodopa and Dl-like agonists, D2-like receptor
agonists such as quinpirole fail to elevate Fos-like immunoreactivity in the 6-OHDA-denervated
striatum (Robertson et al., 1989b;
Robertson et al., 1992; Paul et al., 1992). Instead, quinpirole elevates
Fos-like immunoreactivity
in the ipsilateral globus pallidus. Electrophysiological studies have reported that D2-like receptor activation increases the activity of pallidal neurons and that
6-hydroxydopamine
lesions of the nigrostriatal pathway potentiate this increase (Carlson et al., 1990). The increase in Fos-like
immunoreactivity
in the globus pallidus ipsilateral
to the
6-OHDA-denervated
striatum is consistent with the enhanced
ability of quinpirole to activate pallidal neurons. The small amount
of D2 receptor mRNA in the globus pallidus (Meador-Woodruff
et al., 1989; Mengod et al., 1989) suggests that the excitatory effects
of quinpirole on pallidal neurons are indirect. Given that D2
receptors reside on striatopallidal neurons and function to inhibit
these neurons, it is possible that this pathway is involved in the

Immedmte-Early

Genes as Activity

Markers

247

stimulatory actions of quinpirole on pallidal activity. By inhibiting striatopallidal neurons, quinpirole would presumably decrease
the release of such inhibitory neurotransmitters
as GABA and
enkephalin from striatopallidal terminals resulting in a disinhibition of pallidal neurons. The ability of quinpirole to elevate c-fos
expression in the globus pallidus after 6-OHDA lesions may therefore be a reflection of D2 receptor supersensitivity in the striatum
rather than the globus pallidus. If this is the case, D2-like
receptor-mediated
increases in c-fos expression in the globus
pallidus may serve as an excitatory index of both pallidal and
striatopallidal activity.
Zifl68 (also known as NGFI-A, egr-1 and krox-24) is a transcriptional regulatory factor encoded by the IEG $268 (Changelin et
al., 1989). Like c-fos, ~$268 is considered to be an activity marker
for certain neurons (Worley et al., 1991; Cole et al., 1992). However, unlike c-fos, there is high basal expression of zfl68 in the
striatum. In a recent report, constitutive expression of ~$268
mRNA was detected in medium-sized striatal neurons (Mailleux
et al., 1992). Basal expression of ~$268 appears to be driven by
natural synaptic activity (Worley et al., 1991). ~27268can therefore
be used to measure reductions in neuronal activity. For example,
administration of the selective Dl-like receptor antagonist SCH
23390 reduces basal ~$268 expression in the intact striatum
(Mallieux et al., 1992). Given that Dl receptors in the striatum are
located predominantly on striatonigral neurons, this observation
suggests that SCH 23390 reduces the activity of striatonigral neurons. Reductions in ~$268 expression may therefore be used to
study decreases in striatonigral activity. Furthermore, striatal
~$268 expression is elevated by systemic administration of D2-like
antagonists; whereas its expression is reduced by the D2-like agonist quinpirole (Nguyen et al., 1992; Keefe and Gerfen, 1995). These
changes occur primarily in enkephalin neurons suggesting that
zif268 can also be used to measure both increases and decreases
in the activity of striatopallidal neurons.

4. Members of the Fos and


Jun Family Dimerize to Form the AP- 1 Complex
Fos and Jun are thought to function as transcriptional regulating factors that couple extracellular signals to alterations in cellular phenotype by regulating the expression of specific target genes

242

Robertson

(Morgan and Curran, 1991; Hughes and Dragunow, 1995). In


order to bind to DNA and regulate gene expression, eachfos
family protein must first dimerize with a protein product of
the iun family. Dimerization of Fos and Jun proteins is mediated by hydrophobic interactions between an a-helical domain
containing a heptad repeat of five leucine residues common to
both partners-the
so-called leucine zipper (Gentz et al, 1988;
Turner and Tjian, 1989). Protein products of thefos and @n families can form heterodimers while members of the IWZ family
can also associate with themselves forming
homodimers
(Nakabeppu et al., 1988; Cohen et al., 1989; Zerial et al., 1989;
Nishina et al., 1990). Heterodimers
consisting of members of
the fun andfos families are commonly referred to as Al?-1 complexes which bind to a specific sequence of DNA known as the
AP-1 site (Franz et al., 1988; Rauscher et al., 1988). The leucine
zipper motif permits the formation of a large number of different Al?-1 complexes. For example, proteins encoded by each
member of thefis family (Fos, FosB, Fra-1 and Fra-2) can dimerize with each member of the lun family (Jun, JunB, and JunD)
Accumulating
evidence indicates that the wide variety of
potential dimer combinations serves as a mechanism for fine
transcriptional
regulation.
5. Regulation
of Neuropeptide
Gene Expression by Immediate-Early

Genes

The wide spread inducibility of c-fos expression in the CNS has


led to the search for downstream genes that are regulated by Fos
(Hughes and Dragnow, 1995). AP-l-like binding sites have been
identified in the promoters of genes encoding the neuropeptides
enkephalin, dynorphin, cholecystokinin, and neurotensin, suggesting that their expression may be regulated by Fos (Sonnenberg et
al., 1989; Monstein, 1993; Naranjo et al., 1991, Kislaukis et al., 1988).
Indeed, transient cotransfection studies indicate that Fos and Jun
can enhance proenkephalin
and prodynorphin
expression
(Sonnenberg et al., 1989). However, until recently it had not been
clear as to whether neuropeptide genes were in fact physiological
targets for Fos.
The trldecapeptlde
neurotensm (NT) IS widely distributed
throughout the CNS, where it is thought to function as a classical
neurotransmitter or neuromodulator
(Iversen et al., 1978; Kitabgi

/mmediate-Early

Genes as Actrvity

Markers

243

et al., 1977; Uhl, 1982). A single injection of the prototypical


antipsychotic
haloperidol
produces a dramatic elevation of
neurotensin/neuromedin
N (NT/N) mRNA levels in the dorsolateral striatum suggesting that an increase in synthesis is responsible for the subsequent enhancement of NT concentrations
(Govoni et al., 1980; Merchant et al., 1991; 1992a,b). Several lines
of evidence suggest that c-fos may participate in those intracellular events, responsible for haloperidol-induced
proneurotensin
mRNA expression in the dorsolateral striatum. Several lines of
evidence suggest that c-f& may participate in those intracellular
events responsible for haloperidol-induced
NT/N mRNA expression in the dorsolateral striatum. First, haloperidol dramatically
elevates c-f& mRNA and Fos-like immunoreactivity
(FLI) in the
dorsolateral striatum, with peak increases occurring before those
in NT/N mRNA (Merchant et al., 1992b; Robertson and Fibiger,
1992). Thus, there is a temporal relationship between the c-fos and
NT/N induction. Second, a remarkable correspondence between
the distribution
of haloperidol-induced
c-fos mRNA, FL1 and
NT/N mRNA in the striatum has been noted suggesting that these
increases occur in the same population of neurons (Deutch et al.,
1992; Merchant et al., 1992a, Robertson et al., 1992). Third, an AP1 binding site has been identified in the NT/N promoter that contributes to the inducibility of this gene by nerve growth factor in
PC-12 cells (Kislaulcis and Dobner, 1990). Consistent with this
proposal, we have recently utilized antisense DNA technology to
demonstrate that c-fos induction is necessary for the subsequent
elevation of proneurotensin mRNA in the dorsolateral striatum
by haloperidol (Robertson et al., 1995a). Haloperidol-induced
c-fas
expression was selectively blocked by microinjection
of an
antisense phosphorothioate oligodeoxyribonucleic
(ODN) to this
immediate-early gene into the dorsal striatum. Inhibition of c-fos
expression by the antisense ODN attenuated haloperidol-induced
neurotensin gene expression in the dorsolateral striatum. Selectivity of the antisense effect was confirmed by establishing that
expression of a nontargeted immediate-early gene cc-iun) and neuropeptide (enkephalin), located in striatal neurons that would otherwise have displayed haloperidol-induced
FL1 and c-f& mRNA,
were not altered by the antisense ODN. In this way, we demonstrated that the antisense ODN diminished haloperidol-induced
neurotensin
gene expression by selectively preventing
c-fos
expression.

244

Robertson

6. AfosS as a Chronic

Marker

of Neuronal

Activation

Although immunohistochernical
detection of FL1 has proven
to be a very useful technique for the identification of acute neuronal activation in the CNS, FL1 is limited by the fact that it cannot be used to study neuronal populations that are activated by
chronic stimulation. For instance, chronic administration
of
antipsychotics such as haloperidol or clozapine results in a rapid
desensitization of the acute increases in both c-f& mRNA and FL1
produced by these drugs (Coppers et al., 1995; Merchant et al.,
1995; Sebens et al., 1995). Downregulation
of the c--&s response is
a general phenomenon that has been reported to occur with
repeated exposure to a variety of treatments (Winston et al., 1990,
Hope et al., 1994a; Rosen et al., 1994). In contrast, levels of the IEG
product AFosB are enhanced by chronic exposure to treatments
that acutely elevate Fos (Hope et al., 1994b, Doucet et al., 1996;
Vahid-Ansari et al., 1996). These studies suggest that it may be
possible to use AFosB as a marker for chronic neuronal activation

6.1. AfosB is Produced

by Alternative

Splicing

of fosB

Two different forms of f&B mRNA are generated by alternative splicing of the transcript from a singlefosB gene (Dobrzanski
et al., 1991; Mumberg et al., 1991; Nakabeppu and Nathans, 1991;
Yen et al., 1991). The longer transcript f&B) encodes a protein
338 amino acids in length called FosB, whereas the shorter transcript (AfosB) encodes a truncated form of FosB known as AFosB.
AfosB mRNA is produced by deletion of 140 bases from the fosB
transcript. This deletion shifts the reading frame by a single base,
creating the stop codon TGA. As a result, AFosB is only 237 amino
acids long and lacks the last 101 amino acids found in FosB. Present
within this truncated region is the hepatoproline sequence (amino
acids 257-263) that functions as an activation domain in FosB.
AFosB is therefore a much weaker transcriptional activating factor than FosB but displays prolonged induction kinetics compared
to Fos and FosB.
6.2.

Dopaminergic

Regulation

of A fosB Expression

It is well known that chronic administration of dopaminergic


stimulants that acutely increase c-fos expression leads to a rapid
loss in the ability of these compounds to elevate c-fos expression
in the striatum (Hope et al., 1992; Iadarola et al., 1993; Rosen et

immediate-Early

Genes

as Activity

Markers

245

al., 1994). Despite this desensitization, AP-1 binding remains


elevated suggesting that another member of the fos family is
responsible for the maintenance of transcriptional changes initiated by Fos (Hope et al., 1992). Consistent with this proposal,
repeated administration of the mixed Dl /D2 receptor agonist apomorphine to 6-OHDA-lesioned
rats or of cocaine (an indirect
dopamine agonist) to normal animals, produces a persistent
elevation of FLI, detected with an antibody that recognizes all
known members of thefus family, in the striatum (Zhang et al.,
1992; Hope et al., 1994a). Western blotting and gel-shift experiments indicated that a 35 kDa Fos-related antigen is at least partly
responsible for the prolonged increase in Al?-1 binding produced
by chronic apomorphine or cocaine administration (Pennypacker
et al., 1992; Hope et al., 1994a).
Given that the truncated form of FosB (AFosB) is approx 35 kDa
in size and displays prolonged induction kinetics (Nakabeppu and
Nathans, 1991; Mumberg et al., 1991; Nakabeppu et al., 19931, we
examined the effects of chronic alterations in dopaminergic neurotransmission
on expression of this protein in the striatum
(Doucet et al., 1996). AFosB- and FosB-like immunoreactivity
were
detected using two different affinity-purified
rabbit polyclonal
antibodies (Nakabeppu and Nathans, 1991; Nakabeppu et al.,
1993). One antibody, raised against amino acids 79-131 of the
N-terminus of FosB, recognizes both FosB and AFosB (FosB[N]).
The second antibody, raised against a portion of the C-terminus
of FosB that is missing from AFosB (amino acids 245-3151, recognizes just FosB (FosB[C]) (Nakabeppu
and Nathans, 1991;
Nakabeppu et al., 1993). In a first series of studies, we demonstrated that decreasing striatal D2 receptor stimulation by either
chronic administration of haloperidol or dopaminergic denervation produced a prolonged elevation of FosB-like immunoreactivity detected with the FosB(N) antibody. In contrast, the FosB(C)
antibody failed to demonstrate an increase in FosB-like immunoreactivity after these treatments. These findings were confirmed by
Western blotting that demonstrated a preferential elevation of
AFosB-like proteins. Since the FosB(C) antibody selectively recognizes FosB, these results suggest that chronic elevations in D2
receptor-mediated signaling selectively elevate AFosB expression.
Using retrograde tract tracing techniques to label the major outputs from the striatum, we demonstrated that chronic haloperidol
administration
and destruction of the nigrostriatal
pathway

246

Robertson

selectively increase AFosB levels in striatopallidal neurons. This


observation is consistent with the ability of these treatments to persistently upregulate excitatory transmembrane
signalling in
striatopallidal neurons. In a second series of studies, we investigated the effects of repeated administration of the Dl-like receptor
agonist CY 208-243 (1 mg/kg, injected subcutaneously twice daily
for 5 d) on striatal AFosB levels in rats that had sustained unilateral
lesions of the nigrostriatal pathway. Chronic administration of CY
208-243 produced a dramatic and selective elevation of AFosB
expression in the denervated striatum. Retrograde labelling revealed
that these increases occurred predominantly in striatonigral neurons. This is in line with numerous studies showing that chronic Dl
receptor activation profoundly increases striatonigral gene expression
m the dopaminergically deafferenated striatum. Moreover, our studies suggest that the 35-kDa Fos-related antigen observed by others
after prolonged apomorphine or cocaine administration is actually
AFosB (Pennypacker et al., 1992; Hope et al., 199413).
In summary, our findings indicate that chronic alterations in
dopaminergic neurotransmission produce a prolonged elevation
of AFosB expression in the striatum. Cellular localization studies
indicate that enhanced AFosB expression occurs in neuronal populations that display increases in the expression of genes encoding
a variety of protein classes such as receptors, neuropeptides, and
synthetic enzymes. Inasmuch as these changes appear to be correlated with increases in gene signalling activity, it may be
appropriate to view AFosB as a marker of chronic neuronal activation. If this is the case, it should be possible to use AFosB as a
chronic activity marker in other paradigms in much the same way
that Fos has been used as an acute marker.
References
Badmg, H, Gmty, D D, and Greenberg, M E (1993) Regulation of gene
expression m hrppocampal neurons by drstmct calcmm srgnalmg pathways
Science 260,181-186
Barone, P , Morellr, M , Gcarelh, G , Cozzolmo, A , Deloanna, G , Campanella,
G , and DrChrara, G (1993) Expressron of clfos protem m the experrmental
eprlepsy induced by pllocarpme Synapse 14,1-9
Boyson, S J , McGomgle, P , and Molmoff, P B (1986)Quantrtatrve autoradrographic localrzatron of the Dl and D2 subtypes of dopamme receptors m rat
brain. ] Neuroscl 6,3177-3188
Bulhtt, E (1989)Inductron of c-fos-like protein wrthm the lumbar spinal cord and
thalamus of the rat followmg peripheral strmulatron Braw Res493,391-397

Immediate-Early

Genes as Activity

Markers

247

Carbone, M and Levine, A S. (1990) Oncogenes, antloncogenes, and the regulation of cell growth Trends Endocrlnol Metab 1,248-253
Carlson, J , Bergstrom, D A, Demo, S , and Walters, J R (1990) Nlgrostrlatal
lesions alters neurophysiologlcal responses to selective and nonselective D-l
and D-2 dopamme agonists in rat globus pallldus Synapse 5,83-95
Changelain, P S , Feng, P , King, T C, and Mllbrandt, J, (1983) Structure of the
NGFI-A gene and detection of upsteam sequences responsible for Its transcrlptlonal inductlon by nerve growth factor. Proc Nat1 Acad Scl USA 86,
377-381.
Chase, T N , Mouradlan, M. M , and Engber, T. M. (1993) Motor complications
and the function of striatal efferent systems Neurology 43 (Suppl. 6),523-527
Cohen, D R. and Curran, T (1988) fra-1. serum inducible, cellular lmmedlateearly gene that encodes a Fos-related antigen. Mol Cell Biol. 8,2063-2069
Cohen, D. R , Ferrelra, P C. P , Gentz, R , Franz, Jr., B R , and Curran, T (1989)
The product of a fosrelated gene, Fra-1, binds cooperatively to the AP-1 site
with Jun. transcription
factor AI?-1 IS comprised of multiple protein complexes Genes and Dev 3,173-184
Cole, A J , Abu-Shakra, S., Saffen, D W , Baraban, J M , and Worley, I. (1990a)
Rapid rise in transcription
factor mRNAs m rat brain after electroshock-induced seizures J Neurochem 55,1920-1927
Cole, A J , Bhat, R V , Patt, C , Worley, I?. F ,and Baraban, J. M. (1992) Dl Dopamine receptor actrvatlon of multiple transcription factor genes m rat striatum
J Neuuochem. 58, 1420-1426
Coppens, H. J , Sebens, J B , and Korf, J (1995) Catalepsy, Fos protein, and
dopamme receptor occupancy after long-term haloperldol
treatment
Pharmacol Blochem Behav 51, 175-182.
Curran, T , Miller, A D , Zokas, L , and Verma, I M (1984) Viral and cellular
fos proteins. A comparative analysis. Cell 36, 259-268
Curran, T and Morgan, J I. (1986) Barium modulates c-fos expression and
post-translational modification. Proc Nat1 Acad Su USA 83,8521-8524
Curran, T and Telch, N M. (1982) Candidate product of the FBJ-murme
osteo-sarcomavirus oncogene characterlzatlon of a 55,000dalton phosphoprotem J Vlrol 42,114-X2
Deutch, A. Y , Lee, M C., and Iadarola, M. J (1992) Regionally speclflc effects
of atypical antlpsychotic drugs on strlatal Fos expression the nucleus
accumbensshellasa locusof antipsychotIc actlon Mol. CellNeurosct3,332-341
Dobrzanskl, P, Noguchl, T, Kovary, K., Rlzzo, I., Lazo, P S, and Bravo, R
(1991) Both products of thefosB gene, FosB and Its short form, FosB/SF, are
transcrlptlonal activators m fibroblasts Mol Cell Bzol 11, 5470-5478
Doucet, J I?, Nakabeppu, Y, Bedard, P, Hope, B J., Nestler, E J , Jasmin, B J ,
Chen, J. -S., Iadarola, M J., St-Jean,M., Wigle, N , Blanchet, P , Grondm, R ,
and Robertson, G S (1996)Chronic alterations m dopaminerglc neurotransmisslon produce a persistent elevation of AFosB-hke protein(s) m the stnaturn EUY.J Neurosct 8,365-381
Dragunow, M. and Robertson, H A. (1987)Kindling stlmulatlon induces c-fos
protein(s) m granule cells of the rat dentate gyrus Nature 329,441-442
Dragunow, M , Robertson, G S., Faull, R. L M, Robertson, H A, and Jansen,
K (1990)D2 dopamme receptor antagonists induce Fos and related protems
in rat strlatal neurons. Neurosaence37,287-294.

248

Robertson

Fmkel, M P , Brshs, B 0 ,and Jmkms, P B (1966) Virus mductlon of osteosarcoma m mice Science 151,698-701
Franz Jr, B R , Rauscher III, F J., Josephs, S F , and Curran, T (1988) The Fos
complex and Fos-related antigens recognize sequence elements that contam
AP-1 bindmg sites. Scrence 239,1150-1153.
Gass, I, Herdegen,
T , Bravo, R , and Kiesslmg,
M (1992a) Induction
of
immediate
early gene encoded
protems
in the rat hippocampus
after
brcuculline-induced
seizures-dlfferrentral
expression of KROX-24, FOS and
JUN proteins
Neuroscience 48,315-324.
Gentz, R., Rauscher, F. J III, Abate, C , and Curran, T (1989) Parallel association of Fos and Jun leucine zippers luxtaposes DNA bmdmg domains Scrence
243,1695-1699
Gerfen, C R and Young, W S III (1988) Distribution
of striatomgral
and
strratopallidal
peptidergic
neurons m both patch and matrix compartments
an m situ hybrrdization
hrstochemlstry
and fluorescent
retrograde
tracing
study Brain Res 460, 161-167
Gerfen, C R , Engber, T M., Mahan, L C., Susel, Z , Chase, T N , Monsma, F J ,
Jr, and Sibley, D ,R (1990) D, and D, dopamme
receptor-regulated
gene
expression of striatomgral
and strlatopallidal
neurons Scrence 250,1429-1432.
Gerfen, C R, McGmty, J F and Young, W S III (1991) Dopamme differentially
regulates dynorphin,
substance I, and enkephalm
expression m strlatal neurons in situ hybridization
histochemical
analysis J Neurosct 11,1016-1031
Ghosh, A and Greenberg,
M E (1995) Calcium srgnalmg m neurons
molecular mechanisms
and cellular consequences.
Scrence 268,239-246
Gms, D , Cao, X , Rauscher, F J II I, Cohen, D R , Curran, T, and Sukhatme, V
P (1990) Transcriptional
activation
and repression by Fos are independent
functions.
the C terminus
represses immediate-early
gene expressron
via
CArG elements Mel Cell Bzol 10,4243-4255
Gonzalez, G. A and Montmmy,
M. R (1989) Cyclic AMP stimulates somatostatm
gene transcription
by phosphorylation
of CREB at serine 133 Cell 59,675-680
Govom,
S , Hong, J S , Yang, H Y -T , and Costa, E (1980) Increase of
neurotensm
content elicited
by neuroleptrcs
m nucleus
accumbens
I
Pharmacol Exp Ther 215,413-417
Graybiel, A. M , Moratalla,
R , and Robertson, H A (1990) Amphetamine
and
cocaine induce drugspecific
actlvatron of the c-fos gene m striosorne-matrix
compartments
and hmbrc subdrvrsions
of the striatum
Proc Nat1 Acad Scl
USA 87,6912-6916
Greenberg,
M E. and Zrff, E B (1984) Stimulation
of 3T3 cells induces transcription of the c-fos protooncogene
Nature 311,433-442
Greenberg,
M E , Zrft, E B., and Greene, L A (1986) Stimulation
of neuronal
acetylcholme
receptors induces rapid gene transcription
Science 234,80-83
Gudehithlu,
K P , Neff, N H., and Hadllconstantmou,
M (1933) c-fos and
NGFI-A mRNA of rat retina evidence for hght-induced
augmentation
and a
role for cholinergic
and glutamate receptors
Brarn Res 631,77-82
Harrison,
M B., Wiley, R G , and Wooten, G F (1990) Selective localization
of
strratal Dl receptors to striatomgral
neurons
Brazn Res 528,317-322
Hill, C S , Marau, R , John, S , Wynne, J , Dalton, S , and Trersman, R (1993)
Functional
analysis of a growth factor-responsrve
transcription
factor complex Cell 73,395-406

Immediate-Early

Genes as Activity

Markers

249

Hipskmd,
R. A, Rao, V N , Mueller, C G , Reddy, E S. P., and Norhelm,
A
(1991) Ets-related
protein Elk-l is homologous
to the c-fos regulatory
factor
p62TCF Nature 354,531-534
Hirai, S -I,, Ryseck, R -PI and Mechta, F (1989) Characterization
of lun-D. a
new member of the lun proto-oncogene
family EMBO J 1433-1439.
Hope, B , Kosofsky, B , Hyman, S E , and Nestler, E. J (1992) Regulation
of
immediate-early
gene expression
and AP-1 bmdmg
m the rat nucleus
accumbens by chronic cocaine. Proc Natl. Acad Set USA 89,5764-5768
Hope, 8. T., Kelz, M. B , Duman, R S., and Nestler, E. J (1994a) Chronic electroconvulsive
seizure (ECS) treatment results m expression
of a long-lasting
AP-1 complex
m brain with altered composition
and characteristics
J
Neuroscl 14,4318-4328
Hope, B T., Nye, H E , Kelz, M B., Self, D W , Iadarola, M J , Nakabeppu,
Y ,
Duman, R S , and Nestler, E J. (199417) Induction
of a long-lasting
AP- 1
complex composed of altered Fos-like proteins in brain by chronic cocame
and other chronic treatments. Neuron 13,1235-1244
Hughes, P. and Dragunow,
M. (1995) Induction
of immediate-early
genes and
the control of neurotransmitter-regulated
gene expression within
the nervous system Am Sot Pharm Exp Ther 47,133-178.
Hunt, S P , Pmi, A , and Evan, G (1987) Induction of c-fos-like protein m spmal
cord neurons followmg
sensory stimulation
Nature 328,632-634
Iadarola, M J., Chaung, E J , Yeung, C L., Hoo, Y , Silverthorn,
M , Gu, J , and
Draisci, G (1993) Induction
and suppression
of protooncogenes
m rat striaturn after single or multiple treatments with cocaine or GBR-12909
NINDA
Res Monogr 125,181-211
Iversen, L. L, Iversen, S D , Bloom, F E., Douglas, C , Brown, M., and Vale, W
(1978) Calcium-dependent
release of somatostatm
and neurotensm
m rat
brain. Nature 273, 161-163
Keefe, K and Gerfen, C. R. (1995) Dl-D2
dopamme
receptor
synergy m
striatum
Effects of mtrastriatal
infusions
of dopamme
receptor agonists
and antagonists
on immediateearly gene expression.
Neuroscfence 66(4),
903-913
Kislaukis,
E , Bullock, B , McNeil, S , and Doubner, P R (1988) The rat gene
encoding neurotensm
and neuromedm
N: structure, tissue-specific
expression, and evolution
of exon sequences J Blol Chem 263,4963-4968
Kislaukis,
E and Dobner, P R (1990) Mutually
dependent
response elements
m the cis-regulatory
region of the neurotensin/neuromedin
N gene integrate
enviromental
stimuli in PC12 cells Neuron 4,783-795.
Kitabgi, P , Carraway, R , Van, Retschoten, J., Granier, B, Morgat, J L , Menez,
A, Leeman, S E , and Freychet, P (1977) Neurotensm+
specific bmdmg to
synaptic membranes from rat bram Proc. Nat1 Acad Scl USA 74,1846-1850
Lamph, W W , Wamsley, P , Sassone-Corsi,
I?., and Verma, I. M. (1988) Induction of proto-oncogene
JUN/AP1 by serum and TPA. Nature 334,629-631
Lau, L. F. and Nathans, D (1987) Expression
of a set of growth-regulated
immediate-early
genes in BALB/c3T3
cells: coordmate regulation
with c-fos
or c-myc Proc Nat1 Acad SCI USA 4,1182-1186
Le Gal La Salle, G. and Naquet, R (1990) Audiogemc
seizures evoked m DBA/
2 mice induce c-fos oncogene expression mto subcortical
nucleu BramRes
518,308-312

250

Robertson

Le Mome, C , Normad, E , and Bloch, B (1991) Phenotypical


characterization
of
rat strlatal neurons expressing
the Dl dopamme
receptor gene Proc Nut1
Acad Scl USA 88,4205-4209
Mallieux,
I., Zhang, F., and Vanderhaegen,
J. J (1992) The dopamme
Dl
antagonist SCH 23390 decreases the mRNA levels of the transcription
factor
~$268 (krox-24) m adult rat intact striatum-an
m situ hybridization
study
Neuroscz Lett 147, 182-184
Marars, R , Wynne, J , and Trelsman, R. (1993) The SRF accessory protem Elk-l
contams a growth factor-regulated
transcrrptlonal
activation
domain
Cell
73,381-393.
Meador-Woodruff,
J H , Mansour, A , Bunzow, J R, Van Tol, H H M , Watson,
S J., Jr, and Civelh, 0 (1989) Distribution
of D2 dopamme receptor mRNA
m rat brain. Proc Nat1 Acad Scz USA a&7625-7628
Mengod, G., Martinez-Mir,
I , Vilaro, M T , and Palacros, J M (19891 Locahzatlon of the mRNA for the dopamme D2 receptor in the rat brain by m situ
hybridization
histochemistry
Proc Nat1 Acad Su USA 86,8560-8564
Merchant,
K M., Mrller, M A, Ashleigh, E A, and Dorsa, D M (1991) Haloperidol readily increases the number of neurotensm
mRNA-expressmg
neurons m neostriatum
of the rat bram Brarn Res 540,311-314
Merchant,
K. M , Dobner, I? R , and Dorsa, D M (1992a) Differential
effects of
haloperidol
and clozapme on neurotensm
gene transcription
m rat neostrraturn 1 Neuroscl 12,652-663
Merchant,
K M , Dobie, D J , and Dorsa, D M (1992b) Expression
of the
proneurotensm
gene m the rat bram and its regulation by antipsychotic
drugs
Ann NY Acad Scz 668,54-69
Merchant,
K M , Dobie, D J , F~lloux, F M , Totzke, M , Aravagur,
M , and
Dorsa, D M (1995) Effects of chronic haloperidol
and clozapme treatment
on Neurotensm
and c-fos mRNA m rat neostriatal
subregions
I Pharmacol
Exp Ther 271,460-471
Milbrandt,
J (1987) A nerve growth factor-induced
gene encodes a possible
transcriptional
regulatory
factor Scrence 238,797-799
Monstem, H J (1993) Identlflcatron
of an AP-1 transcription
factor bmdmg site
within the human cholecystokmm
(CCK) promoter
NeuroReport 4,195-197
Montmmy,
M R., Sevarmo, K A, Wagner, J A, Mandel, G , and Goodman, R
H (1986) Identification
of a cyclic-AMP-responsive
element within the rat
somatostatm
gene Proc Nat1 Acad Scz USA 83,6682-6686
Morelli, M , Cozolmo, A , Pmna, A , Fenu, S , Carta, A , and Di-Chiara, G (1993)
L-dopa stimulates c- fos expressron m dopamme denervated strratum by combmed actrvation of D-l and D-2 receptors Bram Res 623,334-336
Morgan, J. I, Cohen, D R , Hempstead,
J. L ,and Curran, T (1987) Mappmg
patterns of c-fos expression m the central nervous system after seizure Sczence
237,192-197
Morgan, J. I and Curran, T (1986) Role of ion flux in the control of c-fos expression Nature 322552-555
Morgan J I and Curran, T (1991) Stimulus-transcription
couplmg m the nervous system mvolvement
of the inducible proto-oncogenesfos
and IUII Ann
Rev Neuroscl 14,421-451
Muller,
R , Bravo, R , Burckhardt,
J , and Curran, T (1984) Induction
of c-fos
gene and protein by growth factors precedes activatron of c-myc Nature 312,
716-720

Immediate-Early

Genes as Actwity

Markers

251

Mumberg, D , Lucibello, F C , Schuermann, M., and Muller, R (1991) Alternative sphcmg of fosB transcripts results in differentially expressed mRNAs
encoding functionally antagonistic proteins Genes and Dev 5,1212-1223
Nakabeppu, Y and Nathans, D (1991) A naturally occurrmg truncated form of
FosB that inhibits Fos/Jun transcriptional activity Cell 64,751-759.
Nakabeppu, Y , Oda, S and Sekiguchi, M. (1993) Proliferative activation of quiescent Rat-1A cells by AFosB Mel Cell Btol 13,4157-4166
Nakabeppu, Y , Ryder, K., and Nathans, D (1988) DNA blmdmg activitres of
three murme Jun proteins stimulation by Fos Cell 55,907-915
Naranlo, J R, Mellstrom, B , Achaval, M , and Sassone-Corsi, P (1991) Molecular pathways of pam Fos/Jun-mediated
activation of a noncanonical AP-1
site m the prodynorphm
gene. Neuron 6,607-617
Nguyen, T V, Kosofsky, B E , Btrnbaum, R , Cohen, B M., and Hyman, S E
(1992) Differential
expression of c-fos and z1p68 m rat striatum after
halopericlol, clozapme and amphetamme
PYOC
Nat1 Acad Scr USA 89,
4270-4274
Nishma, H., Sato, H , Suzuki, T , Sato, N , and Iba, H (1990) Isolation and characterization of fra-2, an additional member of the fos gene family Proc Natl
Acad SCI USA 87,3619-3623
Obseo, J A, Grandas, F , Herrero, M T , and Horowskl,
R (1994) The role of
pulsatile versus contmuous dopamme receptor stimulation for functional
recovery m Parkinsons disease Eur J Neurosct 6,889-897
Page, K J and Everett, B J (1993)Transynaptic mduction of c-fos m basal forebrain, diencephahc and midbrain neurons followmg AMPA-mcluced actlvation of the dorsal and ventral striatum Exp Braln Res.93,399-411
Pan, H S , Penney,
J B , and Young, A B (1985) Ammobutyric acid and benzodiazepme receptor changesinduced by umlateral6-hydroxydopamme lesions
of the medial forebrain bundle ] Neurochem45,1396-1404
Paul, M L , Graybiel, A M , David, J C , and Robertson, H. A (1992) Dl-like
and DZ-like dopamme receptors synergistically activate rotation and c-fos
expression in the dopamme-depleted striatum m a rat model of Parkmsons
disease J Neuroscl 12,3729-3742
Pennypacker, K R., Zhang, W Q , Ye, H., and Hong, J. S (1992)Apomorphme
mductlon
of AP- 1 DNA bmdmg
tion Mol Bum Res 15,151-155

m the rat strlatum

after dopamme

deple-

PO~OVICI, T , Barbm, G , and Ben Ari, Y (1988) Kamic acid-induced seizures increase c-fos-like protem m the hippocampus Etlr J Pkarmucol
150,405-406

Rauscher,F J III, Voulalas, P J , Franza, B R Jr , and Curran, T (1988)Fosand


Jun bmd cooperatively to the AP-1 site reconstitution in wtro Gen Dev. 2,
1687-1699
Robertson, G S , Herrera, D G , Dragunow,
M, and Robertson, H A (1989a)
L-dopa activates c-fos m the striatum ipsilateral to a 6-hydroxydopamme
lesion of the substantia mgra Eur J Pkurmacol 159,99-100
Robertson, G. S , Vmcent, S R , and Fibiger, H C (1990) Striatomgral prolecbon neurons contain Dl dopamme receptor-activated c-fos Brufn Res.523,
288-290

Robertson, G S and Frbiger, H C (1992)Neuroleptics increasec-fosexpression


in the forebrain contrasting effects of haloperidol and clozapme Newosclence
46,315-328

252

Robertson

Robertson, G S , Vincent, S R , and Fibiger, H. C (1992) Dl and D2 dopamme


receptors
differentially
regulate
c-fos expression
m striatonigral
and
striatopallldal
neurons
Neurosczence 49,285-296
Robertson,
G S , Tetzlaff, W., Bedard, A, St-Jean, M , and Wigle, N (1995a)
c-fos mediates antipsychotic-induced
neurotensm
gene expression
in the
rodent striatum
Neuroscience 67,325-344
Robertson,
H. A., Peterson, M. R , Murphy,
K , and Robertson, G S. (1989b)
D,-dopamine
receptor agonists selectively activate striatal c-fos independent
of rotatronal behaviour
Bram Res 503,346-349.
Robertson, L M , Kerppola, T K., Vendrell, M., Luk, D , Smeyne, R J , Bocchiaro,
C , Morgan,
J. I, and Curran, T. (199513) Regulation
of c-fos expression
m
transgemc mice requires multiple
interdependent
transcription
control elements Neuron 14,241-252.
Rosen, J B., Chaung, E , and Iadarola, M J (1994) Differential
induction
of Fos
and Fos-related
antigen followmg
acute and repeated cocame admmistratlon Mel Bratn Res 25,168-172
Ryder, K , Lanahan,
A, Perez-Albuerne,
E , and Nathans, D (1989) Jun-D
a third member
of the Jun gene family. Proc Natl Acad Set USA 86,
1500-1503
Ryder, K , Lau, L. F , and Nathans, D (1988) A gene activated by growth factors
IS related to the oncogene v-lun Proc Nut1 Acad Scz USA 85, 1487-1491
Saffen, D. W , Cole, A J , Worley, P F , Christy, B A, Ryder, K., and Baraban, J
M (1988) Convulsant-induced
increase m transcription
factor messenger
RNAs m rat brain Proc Nat1 Acad Scz USA 85, 7795-7799
Sagar, S M , Sharp, F R , and Curran, T (1988) Expression of c-fos protein in
brain metabohc mapping at the cellular level Science 240,1328-1331
Sassone-Corsl,
I, Slsson, J , and Verma, I M (1988) Transcriptional
autoregulation of the protooncogene
fos. Nature 334,314-319
Schillmg, K L D , Morgan, J I , and Curran, T (1991) Regulation
of afos-lacZ
fusion gene a paradigm
for quantitative
analysis of stimulus-transcription
couplmg
Proc NatL Acad Scr USA 53,5665-5669
Schwanmger,
M, Lux, G., Blume, R, Oetlen, E , Hidaka, H , and Knepel, W
(1993) Membrane
depolarization
and calcium influx induce glucagon gene
transcription
m pancreatic
islet cells through
the cyclic AMP-responsive
element I Bzol Chem 268,5168-5177
Sebens, J B , Koch, T , Ter Horst, G J , and Korf, J (1995) Differential
Fos-protein
mduction
m rat forebrain regions after acute and long-term haloperidol
and
clozapme treatment
Eur J Pharmacol 273,175-182
Shaw, P E , Schroter, H , and Nordheim,
A (1989) The ability of a ternary complex to form over the serum response element correlates with serum mducibihty of the c-fos promoter
Cell 56,563-572
Sheng, M , McFadden,
G , and Greenberg,
M E (1990) Membrane
depolarization and calcium mduce c-fos transcription
via phosphorylation
of transcription factor CREB Neuron 4,571-582.
Sheng, M , Thompson, M. A. and Greenberg, M. E (1991) CREB a Ca2*-regulated
transcription
factor phosphorylated
by calmodulm-dependent
kineses Sczence 252,1427-1430
Smeyne, R J., Curran, T., and Morgan, J. I (1992) Temporal
and spatial expression of a fos-1acZ transgene m the developing
nervous system Mol Bram Res
16,158-162.

Immediate-Early

Genes as Activity

Markers

253

Sonnenberg, J L , MacGregor-Leon, P F., Curran, T , and Morgan, J. I (1989a)


Dynamic alterations occur in the levels and composrtron of transcription factor A-l complexes after seizure Neuron 3,359-365
Sonnenberg, J L , Mitchelmore, C , MacGregor-Leon, P F , Hempstead, J , Morgan, J L , and Curran, T. (1989b) Glutamate receptor agonists increase the
expression of Fos, Fra, and AP-1 DNA binding activity in the mammalian
brain. J Neurosc~Res 24,72-80.
Sonnenberg, J L , Rauscher,F. J III, Morgan, J. I, and Curran, T (1989)Regulation of proenkephalm by Fos and Jun Science246,1622-1625
Sugimoto,T , Sato,K ,Houtam, T , Ueyama,T , and Ikeda,M (1993)On the expression
of Fos-likeprotem rn the subthalamrcnucleusand basalganglia output systems
followrng karnrcacid inlectronsmto the rodent striatum.NeurosczLeft 152,25-28.
Tolle, T R , Casro-Lopes, J M., Cormbra, A, and Zieglgansberger, W (19901
Opiates modify induction of c-fos proto-oncogene in the spinal cord of the
rat followmg noxious stimulation Neurosci Left 111,46-51
Trersman, R (1992) The serum response element Trends Ilzochem Scl 17,
423-426
Trugman, J M and Wooten G F (1987) Selective Dl and D2 dopamme agonists differentially alter basal ganglia glucoseutrlization m rats wrth unilateral 6-hydroxydopaime substantra mgra lesions J Neuroscl 7,2927-2935
Turner, R and Tjran, R (1989) Leucine repeats and an adjacent DNA binding
domain medrate the formation of functional cFos-cJunheterodrmers Science
243,1689-1694
Uhl, G R (1982) Distribution of neurotensm and its receptors in the central
nervous system Ann NY Acad Sci 400,132-149
Vahld-Ansari, F and Robertson, G S. (1996)7-OH-DPAT drfferentrally reverses
clozapme- and haloperidol-induced increasesm Fos-like rmmunoreactivlty
Eur 1 Neuroscl 8,2605-2611
Vendrell, M., Pu~ol,M J , Tusell, J, M , and Serratosa,J (1992)Effect of different
convulsants on calmodulm levels and proto-oncogene c-fos expression m
the central nervous system Mol Bram Res 14, 285-292
Wagner, B J , Hayes, T E , Hoban, C J , and Cochran, B H (1990) The SIF
binding element conferssrs/PDGF inducrbility onto the c-fospromoter. EMBO
J 9,4477-4484
Winston, S M , Hayward, M. D., Nestler, E J , and Duman, R S (1990)Chronic
electroconvulsrve seizuresdown-regulate expressionof the immediate-early
genesc-fosand c-)un m rat cerebral cortex. 1. Neurochem 54,1920-1925
Worley, I F., Chrutry, B A, Nakabeppu, Y , Bhat, R V., Cole, A J , and Baraban,
J M (1991) Constrtutive expression of zif268 m neocortex is regulated by
synaptic activity. Proc NafL Acad. Ser.USA 88,5106-5110
Yen, J , Wisdom, R M , Tratner, I, and Verma, I. M. (1991)An alternatrve spliced
form of FosB ISa negative regulator of transcriptional actrvatron and transformation by Fos proteins Proc Nafl. Acad Scr USA 88,5077-5081
Zerral, M , Toschl, L , Ryseck, R -PI Schuermann, M, Muller, R., and Bravo, R.
(1989) The product of a novel growth factor activated gene, fos-B, interacts
with JUN proteins enhancing then DNA birding activity EMBO J 8,805-813
Zhang, W G , Pennypacker, K R , Ye, H , Merchenthaler, I J , Grimes, L.,
Iadarola, M J, and Hong, J. S (19921 A 35 kD fos-related antigen IS
co-localized with substanceP and dynorphin in striatal neurons. Brmn Res
577,312-317

You might also like