University of Groningen Novel cyclic AMP signalling ... · 4. mAKAP in the mouse brain 5. AKAPs as...

31
University of Groningen Novel cyclic AMP signalling avenues in learning and memory Ostroveanu, A IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record Publication date: 2009 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Ostroveanu, A. (2009). Novel cyclic AMP signalling avenues in learning and memory. s.n. Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum. Download date: 30-06-2021

Transcript of University of Groningen Novel cyclic AMP signalling ... · 4. mAKAP in the mouse brain 5. AKAPs as...

  • University of Groningen

    Novel cyclic AMP signalling avenues in learning and memoryOstroveanu, A

    IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

    Document VersionPublisher's PDF, also known as Version of record

    Publication date:2009

    Link to publication in University of Groningen/UMCG research database

    Citation for published version (APA):Ostroveanu, A. (2009). Novel cyclic AMP signalling avenues in learning and memory. s.n.

    CopyrightOther than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

    Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

    Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons thenumber of authors shown on this cover page is limited to 10 maximum.

    Download date: 30-06-2021

    https://research.rug.nl/en/publications/novel-cyclic-amp-signalling-avenues-in-learning-and-memory(07b1dffb-f0aa-4556-880c-d5182a9adde3).html

  • Chapter 7

    General Discussion

  • Chapter 7

    Content

    1. The role of A-kinase anchoring protein 150 in learning and memory

    2. PKA anchoring to AKAPs in learning and memory

    3. Tools to investigate the function of AKAP(s) signalosome

    4. mAKAP in the mouse brain

    5. AKAPs as new therapeutic targets

    6. The role of Epac in learning and memory

    7. Overall conclusion and future perspectives

    8. Reference

    142

  • General discussion

    1. The role of A-kinase anchoring protein 150 (AKAP150) in

    learning and memory

    Recently, A-kinase anchoring proteins (AKAP) emerged as prototypic coordinators of

    cyclic adenosine monophosphate (cAMP) activated protein kinase (PKA) signaling

    providing high specificity to PKA signaling. The most investigated member of this family

    of proteins is AKAP79/150, a protein expressed in e.g. rodent (AKAP150) and human

    (AKAP79) brain. We showed that AKAP150 is highly expressed in the mouse striatum,

    cerebral cortex, and forebrain regions, while a relatively high expression was found in the

    hippocampus and olfactory bulb of the mouse brain. Low/no expression was observed in

    the cerebellum, hypothalamus, thalamus, and brain stem (Ostroveanu et al., 2007). When

    the AKAP150 staining in the mouse brain is compared to the previously reported

    AKAP150 immunoreactivity pattern in rats (Glantz et al., 1992, Lilly et al., 2005), it

    becomes obvious that the AKAP150 distribution in rat brain is much more homogeneous

    than in the mouse brain. For example, in mice the CA3 area of the hippocampus is very

    densely stained compared to the remainder of the hippocampus (Ostroveanu et al., 2007),

    while the rat hippocampus shows a homogenous staining. Overall, the expression patterns

    of AKAP150 in the mouse and rat brain reveal an abundant presence in brain regions

    involved in learning and memory such as the cortex and hippocampus. Although it is

    difficult to speculate on the role of AKAP150 in the mouse brain on the basis of its

    distribution, these data suggest that AKAP150 may well play a role in learning and

    memory.

    In a study focusing on the distribution of AKAP150 at rat CA1 pyramidal cell postsynaptic

    densities (PSD), it was shown that AKAP150 interacts with components of the excitatory

    PSD, whereas AKAP150 immunoreactivity (IR) was not associated with inhibitory

    synapses (Lilly et al., 2005). In parallel to these findings, AKAP79/150 was identified as a

    constituent of postsynaptic densities (PSD) of excitatory synapses in human cerebral cortex

    (Carr et al., 1992). Moreover, AKAP79/150 is linked to the N- methyl D-aspartate receptor

    (NMDAR) and α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR)

    143

  • Chapter 7

    in the PSD through binding with postsynaptic density-95, discs large, zona occludens-1

    (PDZ) domain, membrane-associated guanylate kinase (MAGUK), scaffold proteins

    postsynaptic density-95 (PSD-95) and synapse-associated protein-97 (SAP97) (Colledge et

    al., 2000). In addition to PKA, AKAP79/150 can also bind protein phosphatase 2B (PP2B)

    and protein kinase C (PKC) (Carr et al., 1992b; Coghlan et al., 1995; Klauck et al., 1996;

    Sik et al., 2000). It was found that this multi-enzyme signaling complex plays an important

    role in coordinating changes in synaptic structure and receptor signaling functions

    underlying synaptic plasticity (Dell'Acqua et al., 2006). For example, AKAP79/150 bound

    PKA was found to be essential for the regulation of AMPA receptor surface expression and

    synaptic plasticity (Rosenmund et al., 1994). Additional evidence that AKAP79/150 could

    be implicated in synaptic transmission arises from the study of Genin and colleagues (2003)

    who showed that AKAP150 mRNA is upregulated during the maintenance phase of long-

    term potentiation (LTP), a mechanism of long-lasting enhancement in synaptic efficacy

    (Genin et al., 2003). A very recent study by Lu et al. (2007) also suggested that AKAP150

    can be involved in synaptic plasticity. In transgenic mice expressing a truncated form of

    AKAP150 which cannot anchor PKA, hippocampal LTP was abolished in 7–12 but not 4-

    week-old mice. From these data they concluded that PKA anchored to AKAP150 critically

    contributes to LTP in the adult hippocampus (Lu et al, 2007). Changes in synaptic plasticity

    are suggested to be the mechanism underlying learning and memory processes (Martin et

    al., 2000; Micheau and Riedel 1999). The findings that AKAP79/150 plays an important

    role in synaptic plasticity, thus, provide additional evidence for the hypothesis that

    AKAP79/150 is important in learning and memory processes. However, knowledge on the

    role of AKAP79/150 in learning and memory remained so far rather limited.

    Initial evidence for a role of AKAPs and PKA anchoring in learning and memory came

    from a study by Moita and colleagues. They blocked PKA anchoring in the lateral

    amygdala of rats and subjected the animals to auditory fear conditioning (Moita et al.,

    2002). Their data showed that inhibition of PKA binding to AKAPs in the rat lateral

    amygdala impairs memory consolidation during auditory fear conditioning (Moita et al.,

    2002). We provided the first direct evidence for a role of AKAP150 in learning and

    memory processes by assessing the expression of AKAP150 in the mouse hippocampus

    144

  • General discussion

    after a single training session in a contextual fear conditioning paradigm. We could show

    for the first time that AKAP150 is upregulated in the mouse hippocampus 6 hours after

    training in this fear conditioning test (Nijholt et al., 2007). The time point of 6 h correspond

    to the late phase of memory consolidation and is critically regulated by cAMP signaling

    pathways in fear motivated associative learning. Moreover, mice exposed to a novel context

    also showed an AKAP150 upregulation in the mouse hippocampus (Nijholt et al., 2007).

    Although we can only speculate on the possible role of the increased expression of

    AKAP150, it might very well be that elevated AKAP150 levels result in a more efficient

    propagation of signals carried by locally generated cAMP (Colledge et al., 2000; Feliciello,

    et al., 1997), which in turn may contribute to processing the exposure to a novel context and

    the consolidation of associative memory. Since AKAP150 levels were only measured 6 h

    after training we cannot exclude that AKAP150 may also be important during other stages

    of the memory process. In general, the upregulation of AKAP150 may be the result of de

    novo protein synthesis or decreased protein degradation.

    Recently several studies provided data on possible mechanism of how AKAP150 could

    modulate learning and memory processes. It was shown that constitutive loss of AKAP150

    in mice modifies excitatory synaptic transmission (Tunquist et al., 2008). This is achieved

    by a deranged localization of PKA in AKAP150 null hippocampal neurons which in turn

    modulates the postsynaptic AMPA receptors. These AKAP150 KO mice also exhibit

    deficits in motor coordination and show memory retrieval impairment in the Morris water

    maze (Tunquist et al., 2008). Hall and colleagues (2007) showed that AKAP150 copurifies

    with L-type Ca2+ channels Cav1.2 in the rat forebrain. Cav1.2 constitutes 80% of the L-type

    channels in the brain (Hell et al, 1993, Moosmang et al., 2005) and is concentrated at

    postsynaptic sites (Hell et al., 1996). It has been shown that hippocampal pyramidal

    neurons express predominantly the Cav1.2 channel (Hell et al., 1993, Davare et al., 2001,

    Sinnegger-Brauns et al., 2004). Interestingly, conditional inactivation of the Cav1.2 gene in

    the mouse hippocampus and neocortex leads to a selective loss of protein synthesis-

    dependent NMDAR-independent Schaffer collateral/CA1 late-phase LTP and these mice

    show a severe impairment in a water-maze spatial-discrimination task (Moosmang et al,

    2005). Although it was shown that Cav1.2 binds several AKAPs in the brain (MAP2B

    145

  • Chapter 7

    (Davare et al., 1999, Hall et al., 2007), AKAP15 (Hall et al., 2007)), AKAP150 proved to

    be critical for PKA-mediated regulation of this channel in the brain (Hall et al., 2007).

    Overall, these data suggest that AKAP150 could be important in learning and memory via

    the regulation of Ca2+ signaling through Cav1.2.

    In addition, Chai et al. found evidence that AKAP150 co-immunoprecipitates with the acid-

    sensing ion channels ASIC1a and ASIC2a (Chai et al., 2007). It has been shown that these

    channels are critically regulated by AKAP150 and calcineurin/PP2B (Chai et al., 2007).

    Interestingly, ASIC1a is abundantly expressed in the amygdala complex and other brain

    regions known to be involved in fear memory, and has been implicated in LTP (Wemmie et

    al., 2002, Chai et al., 2007). Moreover, ASIC1 null mice display deficits in cued and

    contextual fear conditioning (Wemmie et al., 2003), whereas ASIC1a overexpression

    enhances fear-related freezing behavior (Wemmie et al., 2004). Furthermore, inhibition of

    PKA anchoring by Ht-31 induces a decrease in ASIC current amplitude in cultured mouse

    cortical neurons and Chinese hamster ovary (CHO) cells (Chai et al., 2007). Altogether

    these studies suggest that AKAP150 may be able to change memory performance via the

    regulation of ASIC channels.

    Recently, several other AKAPs were reported to be important contributors to learning and

    memory processes. For example, it has been shown that AKAP Yu is involved in the

    formation of long term memory in Pavlovian olfactory learning of Drosophila (Lu et al.,

    2007) and WAVE-1 null-mice (the first AKAP null mouse model) are impaired in spatial

    (Morris water maze) and non-spatial (novel object recognition) memory formation but not

    in emotional learning and memory (passive avoidance) (Soderling et al., 2003).

    In summary, these data provide evidence for an increasing number of AKAPs that are

    involved in learning and memory in different species. Our data, together with future

    findings will lead to the understanding of how AKAPs coordinate the action of signaling

    networks in cognitive processes under physiological and pathological conditions.

    Ultimately this knowledge on the coordination of signaling cascades may lead to the

    development of novel, more fine-tuned, innovative therapeutic strategies to treat cognitive

    dysfunction.

    146

  • General discussion

    2. PKA anchoring to AKAPs in learning and memory

    Besides the role of particular AKAPs in learning and memory, it is at least as important to

    have a good understanding of the role of enzyme binding to these AKAPs. We addressed

    the question whether anchored PKA is critically involved in the different stages

    (acquisition, consolidation, retrieval, and extinction) of contextual fear memory in mice.

    Accordingly, we first injected mice intracerebroventricularly with St-Ht31 (a membrane

    permeable peptide that competes for PKA anchoring) at different time points during the

    memory process. Injecting mice before training with St-Ht31 to affect the acquisition phase

    resulted in impaired associative fear memory 24 h after the training. Similarly, injection of

    St-Ht31 immediately after training (consolidation phase) significantly attenuated

    conditioned fear. The learning deficit observed when St-Ht31 was injected immediately

    after training was similar to the effect of St-Ht31 injected before training. To discriminate

    between acquisition and consolidation, we also performed the retention test 1 h after the

    training with mice that were injected before the training. Interestingly, the performance of

    St-Ht31 injected animals did not differ from the control groups when the retention test was

    performed 1 h after training. The finding that mice which received St-Ht31 before training,

    showed unimpaired freezing 1 h after training but attenuated freezing 24 h after training,

    suggests that PKA anchoring onto AKAPs plays a specific role in the consolidation of

    contextual fear memories but not in acquisition (Nijholt et al., 2008). Moreover, disrupting

    PKA anchoring before the retention test had no effect on memory performance in

    contextual fear conditioning (Nijholt et al., 2008).

    Next, we tested the sub-region specific contribution of PKA anchoring in the hippocampus

    to the consolidation phase of fear memory. Therefore we injected immediately after training

    St-Ht31 locally in the CA1 area of the mouse hippocampus. During the retention test mice

    showed reduced freezing behavior when compared to vehicle or non-injected animals,

    which indicates that disrupting PKA anchoring locally in the CA1 is instrumental in the

    consolidation of fear memory. Our finding that PKA anchoring is important in learning is

    supported by the findings of Nie and colleagues (Nie et al., 2007). Genetically modified

    mice conditionally expressing Ht31 in the forebrain regions show impairments in the spatial

    version of the Morris water maze, a hippocampus-dependent memory task (Nie et al, 2007).

    147

  • Chapter 7

    Although these data showed that PKA anchoring is crucial for memory consolidation, they

    did not provide information on whether type I and/or type II PKA is involved. However,

    most of the AKAPs in the brain tether PKA type II. To date, no RI AKAP was described in

    the brain and only a few dual RI/RII AKAPs were characterized (Huang et al, 1997). Since

    we cannot completely rule out a role for RI anchoring, we injected stearated Super-AKAP-

    IS, in the mouse CA1 area of hippocampus before training mice in contextual fear

    conditioning. Super-AKAP-IS is a PKA anchoring inhibitory peptide that selectively binds

    RIIβ subunits of PKA. This selective anchoring inhibitory peptide also impaired the

    freezing behavior when injected immediately after training. Thus, RIIβ anchoring to

    AKAPs in the CA1 of the mouse hippocampus is crucial and sufficient for the

    consolidation of contextual fear memory.

    Recently Isiegas and colleagues showed that neuronal PKA facilitates the extinction of

    contextual fear (Isiegas et al., 2006). This was found in transgenic mice expressing from

    birth a dominant-negative form of PKA and in mice in which the same PKA dominant-

    negative form was temporarily regulated in brain regions thought to be involved in

    extinction (Isiegas et al., 2006). In our lab, we assessed for the first time the role of PKA

    anchoring in fear extinction. Mice were injected in the CA1 area of the hippocampus with

    stearated Super-AKAP-IS immediately after each extinction trial of contextual fear

    memory. We observed that inhibiting PKA anchoring in the hippocampus promotes the

    extinction of contextual fear conditioning (Nijholt et al., 2008). We thus were the first to

    show that disrupting RIIβ anchoring only in the hippocampus is sufficient to facilitate fear

    extinction. Our data are supported by the finding that temporally transgenic inhibition of

    PKA activity also promotes extinction (Isiegas et al., 2006). More evidence for a role of

    PKA anchoring in memory formation was provided by studies in Drosophila where AKAP-

    bound PKA is required for aversive memory during appetitive memory, a Pavlovian

    olfactory learning task in which an electric shock is used as an aversive unconditioned

    stimulus (Schwaerzel et al., 2007). Unfortunately, St-Ht31 and Super-AKAP-IS do not

    specifically block PKA anchoring to a particular AKAP, and therefore it cannot be

    concluded which AKAP(s) was involved in the impaired consolidation of contextual fear

    memory. We thus determined the amount of PKA anchored to AKAP150 in the dorsal

    148

  • General discussion

    hippocampus after intrahippocampal injection of St-superAKAP-IS. St-superAKAP-IS was

    shown to reduce the amount of PKA anchored to AKAP150 in the dorsal hippocampus

    when compared to vehicle-injected mice.

    It was reported that PKA bound to AKAP150 is crucial for the phosphorylation of GluR1

    AMPA receptor subunit at Ser845 (Colledge et al., 2000, Tavalin et al, 2002). This

    phosphorylation stimulates postsynaptic accumulation of GluR1-containing AMPARs

    during LTP (Esteban et al, 2003; Oh et al, 2006). Previously, it has been shown that

    treatment of hippocampal slices with membrane permeable Ht31 impairs PKA dependent

    hippocampal late-phase LTP (L-LTP) (Huang et al., 2006). Recently, Nie and colleagues

    showed in transgenic mice expressing Ht31 in the hippocampus that a disruption of PKA-

    AKAP interactions does not alter PKA activity or basal synaptic transmission but that only

    long lasting forms of hippocampal LTP are impaired (Nie et al, 2007). Interestingly,

    disruption of PKA anchoring or inhibition of PKA activity in hippocampal neurons leads to

    a PP2B dependent, long-term depression (LTD) -like downregulation of AMPAR currents

    and loss of AMPAR surface expression that likely involves AKAP79/150 (Hoshi et al.,

    2005; Rosenmund et al., 1994; Snyder et al., 2005; Tavalin et al., 2002). Overall, these data

    suggest that AKAP79/150 regulates the balance between PKA and PP2B signaling to

    control AMPAR phosphorylation underlying LTP and LTD in synaptic plasticity. However,

    so far the dynamics of this process are poorly understood. NMDA treatments that induce

    LTD at many synapses simultaneously (chem-LTD) in hippocampal neurons activate

    PP2B/CaN signaling and disrupt the association of AKAP79/150 with PSD-95 and

    cadherins. This leads to a loss of the AKAP from spines and coincides with the removal of

    AMPA receptors from synapses (Gomez et al., 2002; Gorski et al., 2005). In hippocampal

    slices, chem-LTD activation of PP2B/CaN is followed by a persistent redistribution of both

    AKAP79/150 and PKA-RII from postsynaptic membranes to the cytoplasm, without

    PP2B/CaN translocation (Smith et al., 2006). Furthermore, using fluorescence resonance

    energy transfer microscopy in hippocampal neurons, Smith et al. showed that PKA

    anchoring to AKAP79/150 is required for an NMDA receptor-dependent regulated

    cytoplasmic translocation of PKA and AKAP79/150 (Smith et al., 2006).

    In summary, PKA anchoring in the hippocampus is required for both long-lasting forms of

    hippocampal synaptic plasticity, and learning and memory.

    149

  • Chapter 7

    3. Tools to investigate the function of AKAP(s) signalosome

    Besides PKA, there are more AKAP binding partners that participate in learning and

    memory processes. For example, protein phosphatases are already widely acknowledged as

    key molecules in synaptic plasticity and learning and memory. However, the role of protein

    phosphatase anchoring to AKAPs in learning and memory has not been investigated yet.

    The most interesting way to explore the function of any AKAP signalosome is by

    pharmacological studies and/or by genetic manipulation. Although the use of

    pharmacological compounds that block PKA anchoring already yielded fundamental

    insights into the role of AKAP signaling in various physiological systems, inhibitory

    peptides for any other AKAP binding partners are still not available to date. This is most

    likely due to the difficulty of designing such agents and of intracellular delivery, and to the

    expected unspecific action of such peptides.

    In future experiments the impact of the RI isoform-selective anchoring on learning and

    memory processes could be assessed using the RI anchoring disruptor (RIAD) (Carlson et

    al., 2006). To study in greater detail which specific AKAP is involved, it would be

    necessary to develop inhibitors that disrupt the interaction of PKA with one particular

    AKAP or to disrupt the interaction of PKA by introducing site-specific mutations in the

    PKA binding domain of a specific AKAP. Recently, genetically modified mice were

    generated in the AKAP field. It is not surprising that the first complete knockdown of an

    AKAP was AKAP150 (Hall et al., 2007). In addition to the AKAP150 KO mouse, mice in

    which the PKA binding site was removed from the AKAP150 gene were also generated

    (Hall et al., 2007). However, these mice are still not perfect “tools” for investigating the

    role of AKAP150 since the modification occurs also during development and compensatory

    mechanisms cannot be overcome. To define a role for a specific AKAP in learning and

    memory, precise control on the expression of the transgene in space and time is required.

    Another advantage of this spatial inducible system, compared to constitutive deletion of a

    gene or expression of a transgene, is a reduced chance of animal lethality. This is especially

    relevant when the gene of interest plays a crucial role during development or encodes for a

    toxic protein. Already, the tetracycline-dependent system is widely used as a tool for spatio-

    temporal control of transgene expression. Using this system, Isiegas and coworkers

    150

  • General discussion

    generated genetically modified mice that conditionally express Ht31 in forebrain regions.

    Behavioral studies with these Ht31 transgenic mice revealed impairments in spatial learning

    and memory in the Morris water maze as well impairments in long-lasting forms of

    hippocampal LTP (Isiegas et al., 2008).

    In our lab we aimed to generate mice that inducibly express AKAP150 or AKAP150

    mutants only in forebrain regions. In order to explore both the in vitro and in vivo relevance

    of AKAP150 binding to PKA and PP2B, particularly in learning and memory, a strategy

    was developed involving removal of the PKA and PP2B binding site from AKAP150.

    Initially, using a polymerase chain reaction (PCR) based technique we were able to clone

    the mouse AKAP150 protein. Later on, by comparing the mouse AKAP150 sequence with

    the human AKAP79 and rat AKAP150 homologues we could identify the PKA and PP2B

    binding site on mouse AKAP150 and generate several deletion mutants: the PKA binding

    site deletion mutant, the PP2B binding site deletion mutant, and the PKA+PP2B binding

    site deletion mutant (Fig. 1).

    Fig. 1. Left panel. Schematic

    diagram of the AKAP150

    molecule. The PKA and PP2B

    binding site are located near the C-

    terminal end of the protein. The N-

    terminal end of the protein

    contains a binding site for PKC

    and the membrane-targeting

    domain. Right panel. Several mutants were generated: upper - binding site-deleting mutant for PKA, middle -

    binding site-deleting mutant for PP2B, lower - binding site-deleting mutant for both PKA and PP2B

    To generate mice with an AKAP150 overexpression or an AKAP150 mutant

    overexpression we opted for the use of lentiviral vectors to transduce embryonic stem cells.

    These stem cells can then be injected into a mouse blastocyst to create a chimerical

    transgenic animal. Later on, by cross breeding of chimeric transgenic animals, a pure

    transgenic animal can be obtained. These transgenic animals can then be used for

    functional testing to determine the consequences of their altered genetic material (Fig. 2).

    151

  • Chapter 7

    Moreover, by using the lentiviral expression system, various cell lines could be infected in

    addition to test the viability and function of AKAP150 and its mutants in vitro. We

    managed to successfully infect embryonic stem cells with AKAP150 or AKAP150 binding

    site deletion mutants (unpublished data). These results will allow us to move forward in

    creating a transgenic animal expressing inducible AKAP150 binding site deletion mutants

    in the near future. Unquestionably, these transgenic animals can be used in future

    experiments for a better understanding of the role of AKAP150 and anchoring of PKA and

    PP2B to AKAP150 in learning and memory.

    GENE OF INTEREST

    Virus producer cell line

    Producing

    viral particle

    Lentiviral vector

    +

    Gene of interest Overexpress gene of interest

    by infecting the cell line of interest

    In vitro experiments

    +

    Infecting embryonic stem cells

    Select for cells

    expressing

    desired gene

    Inject transformed ES

    cells into blastocyst

    Transgenic animal

    Behavioral

    testing

    Electrophysiology

    Fig. 2. Flowchart of the lentiviral expression system. The genes of interested were cloned in lentiviral expression

    vectors. Viral particles containing the transgene can be use to infect various cell lines. This will allow for in vitro

    experiment to test the function of the wildtype and mutants. Viral particles can also be used to infect embryonic

    stem cells that can be used to generate transgenic animals. The physiological relevance of mutant AKAP150 in

    transgenic animals can be assessed by behavioral testing, in vitro experiments, and electrophysiology.

    4. mAKAP in the mouse brain

    In the last few years, the number of AKAPs that were identified and characterized increased

    considerably. One of these, mAKAP (m of muscular), was found to be predominantly

    152

  • General discussion

    expressed in cardiac and skeletal muscle, but also detected to some extent in the brain

    (Kapiloff et al., 1999). Two alternatively spliced variants of mAKAP were subsequently

    characterized: mAKAPα (preferentially expressed in the brain) and mAKAPβ (abundant in

    cardiac myocytes and skeletal muscle). In addition to their tissue-specific expression,

    mAKAPα and mAKAPβ considerably differ in size: the longer form mAKAPα contains an

    additional 244 amino acid residue N-terminal extension (Michel et al., 2005). Detailed

    knowledge on the localization of mAKAP in the brain remained so far limited. We showed

    that mAKAP is abundantly expressed throughout the entire mouse brain (Ostroveanu et al.,

    2009 submitted). Both immunohistochemistry and Western blotting revealed a high

    expression of mAKAP in the cortex, cerebellum, thalamus, hypothalamus, and

    hippocampus, while the lowest mAKAP expression was detected in the brain stem.

    Moreover, at subcellular levels, mAKAP was abundantly expressed both in perikarya (e.g:

    pyramidal neurons, Purkinje neurons), dendrites (e.g.: apical and basal dendrites of

    hippocampus pyramidal neurons) and to some extent in fibers (e.g: amygdala, cortex). Our

    results are in good agreement with the reported mRNA expression of AKAP100/mAKAP

    in several human brain regions (Kapiloff et. al., 1999; McCartney et al., 1995).

    The function of mAKAP in the brain still remains rather unexplored. Conversely, in the

    heart the function of mAKAP was extensively studied. In the heart, mAKAP is targeted to

    the nuclear envelope through the binding of three spectrin repeat domains, where it forms a

    large macromolecular signaling complex containing several signal transduction molecules,

    including PKA (Kapiloff et al, 1999, Kapiloff et al, 2001), the phosphodiesterase PDE4D3

    (Dodge et al., 2001), the protein phosphatases PP2A and calcineurin (Kapiloff et al, 2001,

    Pare et al., 2005), ryanodine receptors (RyR2) (Kapiloff et al, 2001, Marx et al., 2000,

    Ruehr et al., 2003), the small GTPase Rap1, the guanine exchange factor Epac1 (Dodge-

    Kafka et al., 2005), and the mitogen-activated protein kinases (MAPK) MEK5 and ERK5

    (Dodge-Kafka et al., 2005). Both in the brain and heart, mAKAP can also bind 3’-

    phosphoinositide-dependent kinase 1 (PDK1) and p90 ribosomal S6 kinase (RSK3) (Michel

    et al., 2005). Although in the heart 12 mAKAPβ signalosome components were reported,

    there are probably many more mAKAPβ binding partners.

    The mAKAPα signalosome in the brain has not been characterized yet. It can be expected

    that mAKAPα and mAKAPβ form more or less the same signalosome. Preliminary data

    153

  • Chapter 7

    from our laboratory show that mAKAPα co-immunoprecipitates with PDE4D3 and the

    exchange protein directly activated by cAMP 2 (Epac2) from hippocampal lysates

    (unpublished data). Whether Epac1 is also included in the mAKAPα macromolecular

    complex, still needs to be determined. In addition, preliminary colocalization studies in

    primary cortical neurons confirmed that mAKAP co-localizes with Epac2 and PKA-RIIβ

    (Fig. 3; unpublished data).

    mAKAP PKA-RIIβ Merge

    mAKAP Epac2 Merge

    Fig. 3. mAKAP colocalizes with Epac2 and PKA-RIIβ in primary cortical neurons. Primary cortical neurons from

    E14 were probed with antibodies against mAKAP, Epac2 and PKA-RIIβ.

    Since at the nuclear membrane of cardiomyocytes the mAKAP complex coordinates three

    different cAMP effectors (PKA, PDE4D3 and Epac1), and also calcium and MAPK kinase

    signalling pathways, it is likely that in the brain it has the same function. Since mAKAP

    forms such a large signalosome, it is currently unclear whether there is a single, large

    complex that contains all of the mAKAP binding partners or, alternatively, whether there

    are different mAKAP signalosomes that contain varying combinations of the signaling

    molecules. We detected the highest mAKAP expression presynaptically in brain regions

    such as the central amygdaloid nucleus, supraoptic nucleus, paraventricular hypothalamic

    154

  • General discussion

    nucleus, and suprachiasmatic nucleus (Ostroveanu et al., 2009 submitted). Our finding that

    mAKAP is abundantly expressed, particularly presynaptically, suggests an important role

    for mAKAP in coordinating signal transduction pathways involved in adequate brain

    functioning. This presynaptic expression pattern overlaps remarkably well with the

    expression pattern for corticotrophin releasing factor (CRF) (Pilcher & Joseph, 1984). The

    paralleled distribution of mAKAP with the CRF system suggests that mAKAP may also be

    involved in a variety of functions such as emotion and autonomic responses but also

    memory processes. Furthermore, we found a dramatic decrease in mAKAP expression in

    all brain regions of aged mice. It has been shown that the expression levels of many genes

    related to neuronal signaling, plasticity, and structure are changed in the hypothalamus and

    cortex of the aged mouse brain (Jiang et al., 2001). Several proteins are downregulated in

    the aged brain. For example, the level of PKC β1 is significantly decreased in aged rats as

    assessed by RT-PCR and Western blotting (Yao et al, 1998). Moreover, in brain tissue from

    elderly patients diagnosed with mild cognitive impairments several synaptic proteins (e.g.

    synaptophysin) in the frontal and temporal cortex were shown to be downregulated (Counts

    et al., 2006). Although it is difficult to speculate on the role of mAKAP in the brain on the

    basis of its distribution, it may very well be that mAKAP contributes to changes in brain

    functioning in aged mice and that it plays a role in cognitive processes.

    A growing body of work on the role of AKAPs in the brain and other tissues provides

    evidence that these multifunctional scaffolding proteins facilitate the fidelity of cAMP

    signaling. This tight control of cAMP signaling is clearly important in the maintenance of a

    healthy state, whereas the loss of this regulation might initiate diseases. How mAKAP

    mediates the crosstalk and the integration of different signaling pathways via its

    multimolecular protein complexes and what function it serves in the brain remains,

    however, to be determined.

    5. AKAPs as new therapeutic targets

    Several studies have implicated the AKAP signalosome in ion channel modulation, G-

    protein-coupled receptor desensitization, vesicular secretion, actin cytoskeletal dynamics,

    155

  • Chapter 7

    cell division and gene transcription regulated by cAMP, Ca2+, and lipid second messengers

    (Michel & Scott, 2002). Since many of these cellular processes are altered in human

    disease, understanding the mechanisms of AKAP signaling pathways may aid in the

    development of novel therapeutics. So far traditional treatment in cognitive disorders has

    focused on the manipulation of cell surface protein function. However, pharmacological

    manipulation of cAMP signaling cascades in space and time may ultimately give us more

    control on where and when events occur within the cell. Through careful intervention in the

    coordination of signaling cascades we can gently nudge a cell into a certain direction. By

    targeting anchoring proteins, like AKAPs, we could achieve this subtle intervention.

    6. The role of Epac in learning and memory

    Fairly recently, a novel PKA-independent cAMP effector was discovered and named

    exchange protein directly activated by cAMP (Epac). In independent studies, two variants

    of the Epac protein, Epac1 (also called cAMP-GEF-I) and Epac2 (also called cAMP-GEF-

    II), were characterized (de Rooij et al., 1998; Kawasaki et al., 1998). Both Epacs function

    as guanine nucleotide-exchange factors (GEFs) that specifically activate Rap GTPases

    (Rap1 and Rap2) upon binding to cAMP. Although Epac proteins have been found to

    control key cellular processes, including cellular calcium handling, integrin-mediated cell

    adhesion, gene expression, cardiac hypertrophy, inflammation, and exocytosis (Pereira et

    al., 2007; Oestreich et al., 2007; Hucho et al., 2005; Rangarajan et al., 2003; Lotfi et al.,

    2006; Kang et al., 2003; Morel et al., 2005), their role in the brain just started to be

    investigated. Since cAMP signaling is established to be of critical importance in learning

    and memory, a potential role for PKA-independent cAMP signaling through Epac proteins

    in the process of learning and memory may be expected. Recently, Epac has been linked to

    synaptic transmission and neuronal excitability (Zhong & Zucker, 2005), was shown to

    modulate membrane potentials in cultured cerebellar neurons, thereby potentially regulating

    cellular excitability (Ster et al., 2007), and to enhance neurotransmitter release at excitatory

    synapses (Gekel & Neher, 2008). Very recently it has been shown that Epac is involved in

    both LTP (Gelinas et al, 2008) and LTD (Ster et al., 2009) in the mouse hippocampus.

    156

  • General discussion

    Since both changes in LTP and LTD that affect synaptic transmission in the hippocampus

    are thought to be involved in memory formation, these data suggest furthermore the

    involvement of Epac in learning and memory processes.

    We showed that intrahippocampal delivery of 8-pCPT-2’O-Me-cAMP, a compound that

    specifically activates Epac but not PKA, improves fear memory retrieval in contextual fear

    conditioning whereas acquisition and consolidation were not affected (Ostroveanu et al.,

    2009 submitted). The retrieval enhancing effect of the Epac activator was even more

    prominent in the passive avoidance test, a behavioral paradigm based on associative

    emotional learning similar to fear conditioning. Since the performance of the mice in the

    retention tests may be influenced by the level of anxiety the animal experiences, we tested

    the effect of 8-pCPT-2’O-Me-cAMP on anxiety behavior in an elevated plus maze.

    Intrahippocampal 8-pCPT-2’O-Me-cAMP injection before the elevated plus maze had no

    effect on anxiety. Therefore, the effect of Epac activation in the fear-motivated learning

    tasks can be solely ascribed to enhanced memory retrieval of the association between the

    electric footshock and the context (Ostroveanu et al., 2009 submitted). Ouyang and

    colleagues also recently reported a role for Epac signaling in memory retrieval (Ouyang et

    al., 2008). In their study, the memory retrieval impairment observed in dopamine-beta-

    hydroxylase deficient mice could be rescued by intrahippocampal (i.h.) injection of a

    selective PKA activator together with a selective Epac activator, whereas injection of one

    of the activators alone did not overcome the retrieval deficit (Ouyang et al., 2008).

    Interestingly, when cAMP agonists were infused into the dorsal hippocampus of wild-type

    mice 30 min before testing retrieval, no significant effects on retrieval were observed when

    the agonists were infused separately or in combination (Ouyang et al., 2008). The lack of

    any effect on memory retrieval in the wild type mice is in contradiction with our findings

    and may depend on the time of agonist delivery. Ouyang and colleagues performed the

    injections 30 minutes before the retrieval, whereas in our experiments, Epac agonist was

    injected into the CA1 are of the hippocampus 10 minutes before the memory test.

    It has been shown that 8-pCPT-2’O-Me-cAMP activates both Epac1 and Epac2. Therefore,

    the enhanced memory retrieval effect cannot be attributed to a particular Epac. We showed

    157

  • Chapter 7

    that Epac2 is abundantly expressed throughout the whole mouse brain, whereas only low

    levels of Epac1 were detected. Using semi-quantitative RT-PCR for Epac1 and Epac2 with

    mRNA isolated from the hippocampus, Epac2 mRNA could be detected much earlier as

    Epac1 mRNA, pointing towards much higher Epac2 than Epac1 mRNA levels in the

    hippocampus (Ostroveanu et al., 2009 submitted). Our data are consistent with a previous

    study from Kawasaki et al. who also reported a high expression of Epac2 in the rat brain

    whereas Epac1 was barely detectable (Kawasaki et al., 1998). To date, no Epac-specific

    inhibitors are available. Therefore, we established a protocol for in vivo lipid mediated

    siRNA gene silencing in the mouse brain to investigate the role of Epac2 in learning and

    memory. Local Epac2 silencing in the CA1 area of the hippocampus led to impaired

    memory retrieval 3 days after conditioning whereas Epac2 silencing during the retention

    test 17 days after conditioning had no effect on memory retrieval indicating a time-limited

    function of Epac2 signaling after conditioning. The finding that Epac2 is abundantly

    expressed in the mouse brain together with the finding that Epac2 silencing impairs

    memory retrieval, suggests an important role for Epac2 in learning and memory. In our

    experiments we could not completely exclude a role for Epac1 in these processes. Both

    specific-Epac inhibitors and/or transgenic or knock-out mice will provide further evidence

    on the role of Epac1 and Epac2 in cognitive processes.

    Interestingly, expression levels of both Epac1 and Epac2 were reported to be altered in the

    frontal cortex and hippocampal regions of brains showing Alzheimer’s pathology (McPhee

    et al., 2005). Alzheimer disease (AD) is a neurodegenerative disease characterized by the

    formation of β-amyloid plaques and neurofibrillary tangles. It has been shown that

    amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic

    plasticity and memory (Shankar et al., 2008). Moreover, in AD, neural degradation occurs

    primarily in neurons involved in memory storage and retrieval (Arshavsky, 2006). So far

    the relationship between changes in Epac expression and Alzheimer’s pathology remain

    unclear, but it could very well be that Epac plays a role in the cognitive decline such as

    observed in AD. This is also supported by the finding that the Epac2 levels are decreased in

    AD. We have shown in our studies that activation of Epac in the mouse hippocampus

    significantly and specifically improves the retrieval of fear memory. Considering both the

    158

  • General discussion

    lack and the need of drugs proven to be effective in improving memory retrieval, the

    specific facilitating effect of Epac activation on retrieval we observed, is of particular

    interest and deserves further research into the role of Epac signaling in cognitive processes

    under physiological and pathological conditions.

    Recent findings in several physiological systems acknowledged Epac as coordinator of

    specific cAMP signals independent of PKA. Interestingly, it appears that in part, signals

    carried through Epac are spatially and temporally compartmentalized. For example, in the

    heart mAKAP is able to target Epac1 (Dodge-Kafka et al., 2005), while in the brain

    AKAP150 binds both Epac1 and Epac2 (Nijholt et al, 2008). Thus, the spatio-temporal

    integration of cAMP pathways via Epac, PKA, AKAPs and PDEs dramatically increases

    the complexity and, consequently, the possible readouts of cAMP signalling. Moreover,

    since both Epac and PKA are ubiquitously expressed, an increase in cAMP levels will lead

    to the activation of both effectors which in turn can execute independent responses or

    develop a cross-talk effect. For example, it has been reported that PKA and Epac can exert

    opposing effects on the regulation of the PKB/AKT pathway. While PKA suppresses PKB

    phosphorylation and activity, activation of Epac leads to increased PKB phosphorylation

    (Mei et al., 2002; Nijholt et al., 2008). Interestingly, we recently showed that PKA and

    Epac-mediated PKB/Akt phosphorylation is coordinated in neurons by AKAP150 (Nijholt

    et al., 2008). In addition to the opposing effects, PKA and Epac may also work

    synergistically in other systems. For example both the stimulation of neurotensin (Li et al.,

    2007) and the attenuation of cAMP signaling through phosphodiesterases depend on the

    activation of Epac and PKA (Dodge-Kafka et al., 2005).

    Future research will lead to a better understanding of the physiological roles of both Epac

    isoforms. Eventually, this may lead to a more detailed knowledge of the distinct role that

    individual Epacs play in shaping cAMP signalling in the mammalian brain. This in turn

    could result in the development of new strategies for selective pharmacologic manipulation

    of the different cAMP signaling systems in cognitive processes under physiological and

    pathological conditions.

    159

  • Chapter 7

    7. Overall conclusion and future perspectives

    In the current thesis we aimed to investigate the role of two recently discovered, cAMP

    signaling avenues, via A-kinase anchoring proteins (AKAPs) and via exchange protein

    activated by cAMP (Epac), in memory processes. A large number of publications

    established an instrumental role of cAMP signaling in learning and memory processes.

    With the discovery of both AKAPs and Epac, cAMP research has undergone a revival and

    there grew a general awareness that cAMP signaling is much more complex than was

    initially believed. Accordingly, it has become apparent that AKAPs and Epac might

    mediate a more fine-tuned level of organization for cAMP second messenger systems. Our

    results yield important insights in the role of AKAPs, compartmentalized cAMP signaling

    and Epac in the different stages of learning and memory.

    It was previously believed that anchoring proteins in the brain were only scaffolds for the

    signaling enzymes involved in cognitive processes. Nevertheless, recent data, including our

    own, show a completely new role for AKAPs as dynamic and active contributors to the

    molecular machinery of learning and memory. With the increased number of anchoring

    proteins discovered in the brain, careful analysis on the individual role and contribution of

    AKAPs in learning and memory will continue to be an important part of future research.

    Unquestionably, further analysis of AKAP macromolecular complexes role in learning and

    memory will be forthcoming from genetic approaches involving transgenic mice.

    Genetically modified mice expressing truncated AKAP molecules that hinder tethering of

    signaling enzymes, or mice lacking a particular AKAP, will permit a more precise

    dissection of particular AKAP-PKA-signaling enzymes/complexes in vivo. These mice will

    not only provide insight into the mechanisms of AKAP functions but may yield genetic

    models of specific diseases.

    In this thesis, we could also show that cAMP signals independent of PKA pathways are

    essential events in learning and memory processes. Consequently, compelling evidence

    points towards an important role for Epac in these processes. However, the mechanism by

    160

  • General discussion

    which Epac activation affects learning and memory is still unknown. Understanding how

    exactly cAMP signals are coordinated will bring light on the elusive process of learning and

    memory and memory retrieval in particular.

    cAMP signaling was shown to be crucial in cognitive deficits and may consequently be a

    promising target for therapeutic agents. Clearly, the ultimate goal of research on learning

    and memory is translating knowledge from the laboratory to clinical applications. Thus, in

    addition to the fundamental significance of understanding cAMP signaling cascades, our

    research opens up new routes for the development of new therapeutic strategies for

    memory-associated disorders and pathologies.

    8. Reference

    Arshavsky, Y.I. (2006). Alzheimer's disease, brain immune privilege and memory: a hypothesis.

    Journal of Neural Transmission 11, 1697-1707.

    Carlson, C.R., Lygren, B., Berge, T., Hoshi, N., Wong, W., Taskén, K., & Scott, J.D. (2006).

    Delineation of type I protein kinase A-selective signaling events using an RI anchoring disruptor.

    Journal of Biological Chemistry 281, 21535-21545.

    Carr, D. W., Hausken, Z. E., Fraser, I. D. C., Stofko-Hahn, R. E., & Scott, J. D. (1992). Association

    of the Type I1 CAMP-dependent Protein Kinase with a Human Thyroid RII-anchoring Protein.

    Cloning and characterization of the RII-binding domain. Journal of Biological Chemistry 267, 13376-

    13382.

    Carr, D.W., Stofko-Hahn, R.E., Fraser, I.D.C., Cone, R.D., & Scott, J.D. (1992). Localization of the

    cAMP-dependent Protein Kinase to the Postsynaptic Densities by A-Kinase Anchoring Proteins.

    Journal of Biological Chemistry 24, 16816-16823.

    Chai, S., Li, M., Lan, J., Xiong, Z.G., Saugstad, J.A., & Simon, R.P. (2007). A kinase-anchoring

    protein 150 and calcineurin are involved in regulation of acid-sensing ion channels ASIC1a and

    ASIC2a. Journal of Biological Chemistry 31, 22668-22677.

    161

    http://www.ncbi.nlm.nih.gov/pubmed/16932992?ordinalpos=6&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSum

  • Chapter 7

    Coghlan, V.M., Perrino, B.A., Howard, M., Langeberg, L.K., Hicks, J.B., Gallatin, W.M., & Scott,

    J.D. (1995). Association of protein kinase A and protein phosphatase 2B with a common anchoring

    protein. Science 5194, 108-111.

    Colledge, M., Dean, R.A., Scott, G.K., Langeberg, L.K., Huganir, R.L., & Scott, J.D. (2000).

    Targeting of PKA to glutamate receptors through a MAGUK-AKAP complex. Neuron 27, 107-119.

    Counts, S.E., Nadeem, M., Lad, S.P., Wuu, J., & Mufson, E.J. (2006). Differential expression of

    synaptic proteins in the frontal and temporal cortex of elderly subjects with mild cognitive

    impairment. Journal of Neuropathology and Experimental Neurology 6, 592-601.

    Davare, M.A., Dong, F., Rubin, C.S., & Hell, J.W. (1999). The A-kinase anchor protein MAP2B and

    cAMP-dependent protein kinase are associated with class C L-type calcium channels in neurons.

    Journal of Biological Chemistry 42, 30280-30287.

    Davare, M.A., Avdonin, V., Hall, D.D., Peden, E.M., Burette, A., Weinberg, R.J., Horne, M.C.,

    Hoshi, T., & Hell, J.W. (2001). A beta2 adrenergic receptor signaling complex assembled with the

    Ca2+ channel Cav1.2. Science 5527, 98-101.

    de Rooij, J., Zwartkruis, F.J., Verheijen, M.H., Cool, R.H., Nijman, S.M., Wittinghofer, A., & Bos,

    J.L. (1998). Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP.

    Nature 396, 474-477.

    Dell'Acqua, M.L., Smith, K.E., Gorski, J.A., Horne, E.A., Gibson, E.S., & Gomez, L.L. (2006).

    Regulation of neuronal PKA signaling through AKAP targeting dynamics. European Journal of Cell

    Biology 85, 627-633.

    Dodge, K. L., Khouangsathiene, S., Kapiloff, M. S., Mouton, R., Hill, E. V., Houslay, M. D.,

    Langeberg, L. K., & Scott, J. D. (2001). mAKAP assembles a protein kinase A/PDE4

    phosphodiesterase cAMP signaling module. EMBO Journal 20, 1921-1930.

    Dodge-Kafka, K.L., Soughayer, J., Pare, G.C., Carlisle Michel, J.J., Langeberg, L.K., Kapiloff, M.S.,

    & Scott, J.D. (2005). The protein kinase A anchoring protein mAKAP coordinates two integrated

    cAMP effector pathways. Nature 437, 574-578.

    Esteban, J.A., Shi, S.H., Wilson, C., Nuriya, M., Huganir, & R.L., Malinow, R. (2003). PKA

    phosphorylation of AMPAR subunits controls synaptic trafficking underlying plasticity. Nature

    162

    http://www.ncbi.nlm.nih.gov/pubmed/7528941?ordinalpos=33&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/pubmed/7528941?ordinalpos=33&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSum

  • General discussion

    Neuroscience 6, 136-143.

    Feliciello A., Li, Y., Avvedimento, E.V., Gottesman, M. E., &Rubin, C. S. (1997). A-kinase anchor

    protein 75 increases the rate and magnitude of cAMP signaling to the nucleus. Current Biology, 7,

    1011-1014.

    Gekel, I., & Neher, E. (2008). Application of an Epac activator enhances neurotransmitter release at

    excitatory central synapses. Journal of Neuroscience 28, 7991-8002.

    Gelinas, J.N., Banko, J.L., Peters, M.M., Klann, E., Weeber, E.J., & Nguyen, P.V. (2008). Activation

    of exchange protein activated by cyclic-AMP enhances long-lasting synaptic potentiation in the

    hippocampus. Learning and Memory 15, 403-411.

    Genin, A., French, P., Doyere, V., Davis, S., Errington, M. L., Maroun, M., Stean, T., Truchet, B.,

    Webber, M., Wills, T., Richter-Levin, G., Sanger, G., Hunt, S. P., Mallet, J., Laroche, S., Bliss, T.V.,

    & O'Connor, V. (2003). LTP but not seizure is associated with up-regulation of AKAP-150.

    European Journal of Neuroscience, 17, 331-340.

    Glantz, S.B., Amat, J.A., & Rubin, C.S., (1992). cAMP signaling in neurons: patterns of neuronal

    expression and intracellular localization for a novel protein, AKAP150, that anchors the regulatory

    subunit of cAMP-dependent protein kinase II beta. Molecular Biology of the Cell 3, 1215-1228.

    Gomez, L.L., Alam, S., Smith, K.E., Horne, E., & Dell'Acqua, M.L. (2002). Regulation of A-kinase

    anchoring protein 79/150-cAMP-dependent protein kinase postsynaptic targeting by NMDA receptor

    activation of calcineurin and remodeling of dendritic actin. Journal of Neuroscience 16, 7027-7044.

    Gorski, J.A., Gomez, L.L., Scott, J.D., & Dell'Acqua, M.L. (2005) Association of an A-kinase-

    anchoring protein signaling scaffold with cadherin adhesion molecules in neurons and epithelial cells.

    Molecular Biology of the Cell 8, 3574-3590.

    Hall, D.D., Davare, M.A., Shi, M., Allen, M.L., Weisenhaus, M., McKnight, G.S., & Hell, J.W.

    (2007). Critical role of cAMP-dependent protein kinase anchoring to the L-type calcium channel

    Cav1.2 via A-kinase anchor protein 150 in neurons. Biochemistry 6, 1635-1646.

    Hell, J.W., Westenbroek, R.E., Warner, C., Ahlijanian, M.K., Prystay, W., Gilbert, M.M., Snutch,

    T.P., & Catterall, W.A. (1993). Identification and differential subcellular localization of the neuronal

    class C and class D L-type calcium channel alpha 1 subunits. The Journal of Cell Biology 4, 949-962.

    163

  • Chapter 7

    Hell, J.W., Westenbroek, R.E., Breeze, L.J., Wang, K.K., Chavkin, C., & Catterall, W.A. (1996) N-

    methyl-D-aspartate receptor-induced proteolytic conversion of postsynaptic class C L-type calcium

    channels in hippocampal neurons. Proceedings of the National Academy of Sciences United States of

    America 8, 3362-3367.

    Hoshi, N., Langeberg, L.K., & Scott, J.D., (2005). Distinct enzyme combinations in AKAP signaling

    complexes permit functional diversity. Nature Cell Biology 7, 1066-1073.

    Huang, L.J., Durick, K., Weiner, J.A., Chun, J., & Taylor, S.S. (1997). Identification of a novel

    protein kinase A anchoring protein that binds both type I and type II regulatory subunits. Journal of

    Biological Chemistry 12, 8057-8064.

    Huang, L.J., Durick, K., Weiner, J.A., Chun, J., & Taylor, S.S. (1997). D-AKAP2, a novel protein

    kinase A anchoring protein with a putative RGS domain. Proceedings of the National Academy of

    Sciences United States of America 21, 11184-11189. Huang, T., McDonough, C. B., & Abel T. (2006). Compartmentalized PKA signaling events are

    required for synaptic tagging and capture during hippocampal late-phase long-term potentiation.

    European Journal of Cell Biology, 85, 635-642.

    Hucho, T.B., Dina, O.A., & Levine, J.D. (2005). Epac mediates a cAMP-to-PKC signaling in

    inflammatory pain: an isolectin B4(+) neuron-specific mechanism. Journal of Neuroscience 25, 6119-

    6126.

    Isiegas, C., Park, A., Kandel, E.R., Abel, T., & Lattal, K.M. (2006). Transgenic inhibition of neuronal

    protein kinase A activity facilitates fear extinction. Journal of Neuroscience 26, 12700-12707.

    Isiegas, C., McDonough, C., Huang, T., Havekes, R., Fabian, S., Wu, L.J., Xu, H., Zhao, M.G., Kim,

    J.I., Lee, Y.S., Lee, H.R., Ko, H.G., Lee, N., Choi, S.L., Lee, J.S., Son, H., Zhuo, M., Kaang, B.K., &

    Abel, T. (2008). A novel conditional genetic system reveals that increasing neuronal cAMP enhances

    memory and retrieval. Journal of Neuroscience 24, 6220-6230.

    Jiang, C.H., Tsien, J.Z., Schultz, P.G.,& Hu, Y. (2001). The effects of aging on gene expression in the

    hypothalamus and cortex of mice. Proceedings of the National Academy of Sciences United States of

    America 4, 1930-1934.

    Kang, G., Joseph, J.W., Chepurny, O.G., Monaco, M., Wheeler, M.B., Bos, J.L., Schwede, F.,

    Genieser, H.G., & Holz, G.G. (2003). Epac-selective cAMP analog 8-pCPT-2'-O-Me-cAMP as a

    164

    http://www.ncbi.nlm.nih.gov/pubmed/18550764?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/pubmed/18550764?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/pubmed/11172053?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/pubmed/11172053?ordinalpos=2&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Kang%20G%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Joseph%20JW%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Chepurny%20OG%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Monaco%20M%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Wheeler%20MB%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Bos%20JL%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Schwede%20F%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Genieser%20HG%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Holz%20GG%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlus

  • General discussion

    stimulus for Ca2+-induced Ca2+ release and exocytosis in pancreatic beta-cells. Journal of Biological

    Chemistry 10, 8279-8285.

    Kapiloff, M.S., Schillace, R.V., Westphal, A.M., & Scott, J.D. (1999). mAKAP: an A-kinase

    anchoring protein targeted to the nuclear membrane of differentiated myocytes. Journal of Cell

    Science 112, 2725-2736.

    Kapiloff, M.S., Jackson, N., & Airhart, N. (2001). mAKAP and the ryanodine receptor are part of a

    multi-component signaling complex on the cardiomyocyte nuclear envelope. Journal of Cell Science

    17, 3167-3176.

    Kapiloff M.S., Schillace R.V., Westphal A.M., & Scott J.D. (1999). mAKAP: an A-kinase anchoring

    protein targeted to the nuclear membrane of differentiated myocytes. Journal of Cell Science 114,

    3167-3176.

    Kawasaki, H., Springett, G.M., Mochizuki, N., Toki, S., Nakaya, M., Matsuda, M., Housman, D.E., &

    Graybiel, A.M. (1998). A family of cAMP-binding proteins that directly activate Rap1. Science 282,

    2275-2279.

    Klauck, T.M., Faux, M.C., Labudda, K., Langeberg, L.K., Jaken, S., & Scott, J.D. (1996).

    Coordination of three signaling enzymes by AKAP79, a mammalian scaffold protein. Science 271,

    1589-1592.

    Li, J., O'Connor, K.L., Cheng, X., Mei, F.C., Uchida, T., Townsend, C.M. Jr., & Evers, B.M. (2007).

    Cyclic adenosine 5'-monophosphate-stimulated neurotensin secretion is mediated through Rap1

    downstream of both Epac and protein kinase A signaling pathways. Molecular Endocrinology 1, 159-

    171.

    Lilly, S.M., Alvarez, F.J., & Tietz, E.I., (2005). Synaptic and subcellular localization of A-kinase

    anchoring protein 150 in rat hippocampal CA1 pyramidal cells: co-localization with excitatory

    synaptic markers. Neuroscience 1, 155-163.

    Lotfi, S., Li, Z., Sun, J., Zuo, Y., Lam, P.P., Kang, Y., Rahimi, M., Islam, D., Wang, P., Gaisano,

    H.Y., & Jin, T. (2006). Role of the exchange protein directly activated by cyclic adenosine 5'-

    monophosphate (Epac) pathway in regulating proglucagon gene expression in intestinal endocrine L

    cells. Endocrinology 8, 3727-3736.

    165

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Graybiel%20AM%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/pubmed/16644915?ordinalpos=19&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/pubmed/16644915?ordinalpos=19&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/pubmed/16644915?ordinalpos=19&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSum

  • Chapter 7

    Lu, Y., Allen, M., Halt, A.R., Weisenhaus, M., Dallapiazza, R.F., Hall, D.D., Usachev, Y.M.,

    McKnight, G.S., & Hell, J.W. (2007) Age-dependent requirement of AKAP150-anchored PKA and

    GluR2-lacking AMPA receptors in LTP. EMBO Journal 26, 4879-4890.

    Martin, S.J., Grimwood, P.D., & Morris, R.G. (2000). Synaptic plasticity and memory: an evaluation

    of the hypothesis. Annual Review of Neuroscience 2, 649-711.

    Marx, S.O., Reiken, S., Hisamatsu, Y., Jayaraman, T., Burkhoff, D., Rosemblit, N., & Marks, A. R.

    (2000). PKA phosphorylation dissociates FKBP12.6 from the calcium release channel (ryanodine

    receptor): defective regulation in failing hearts. Cell 101, 365-376.

    McCartney, S., Little, B.M., Langeberg, L.K., & Scott, J.D. (1995). Cloning and characterization of

    A-kinase anchor protein 100 (AKAP100). A protein that targets A-kinase to the sarcoplasmic

    reticulum. Journal of Biological Chemistry 270, 9327-9333.

    McPhee, I., Gibson, L.C., Kewney, J., Darroch, C., Stevens, P.A., Spinks, D., Cooreman, A., &

    MacKenzie, S.J. (2005) Cyclic nucleotide signaling: a molecular approach to drug discovery for

    Alzheimer's disease. Biochemical Society Transactions 33, 1330-1332.

    Mei, F.C., Qiao, J., Tsygankova, O.M., Meinkoth, J.L., Quilliam, L.A., & Cheng, X. (2002).

    Differential signaling of cyclic AMP: opposing effects of exchange protein directly activated by

    cyclic AMP and cAMP-dependent protein kinase on protein kinase B activation. Journal of

    Biological Chemistry 13, 11497-11504.

    Micheau, J., & Riedel, G. (1999). Protein kinases: which one is the memory molecule? Cellular and

    Molecular Life Sciences 4, 534-548.

    Michel J., Townley, I.K., Dodge-Kafka, K.L., Zhang, F., Kapiloff, M.S., & Scott, J.D. (2005). Spatial

    restriction of PDK1 activation cascades by anchoring to mAKAPα. Molecular Cell 20, 661-672.

    Michel, J.J., & Scott, J.D. (2002). AKAP mediated signal transduction. Annual Review of

    Pharmacology and Toxicology 42, 235-257.

    Moita, M.A., Lamprecht, R., Nader, K., & LeDoux, J.E., (2002). A-kinase anchoring proteins in

    amygdala are involved in auditory fear memory. Nature Neuroscience 5, 837-838.

    Moosmang, S., Haider, N., Klugbauer, N., Adelsberger, H., Langwieser, N., Müller, J., Stiess, M.,

    Marais, E., Schulla, V., Lacinova, L., Goebbels, S., Nave, K.A., Storm, D.R., Hofmann, F., &

    166

    http://www.ncbi.nlm.nih.gov/pubmed/10845078?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/pubmed/10845078?ordinalpos=4&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Mei%20FC%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Qiao%20J%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Tsygankova%20OM%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Meinkoth%20JL%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Quilliam%20LA%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Cheng%20X%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/pubmed/11807172?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSum

  • General discussion

    Kleppisch, T.(2005). Role of hippocampal Cav1.2 Ca2+ channels in NMDA receptor-independent

    synaptic plasticity and spatial memory. Journal of Neuroscience 43, 9883-9892.

    Nie, T., McDonough, C.B., Huang, T., Nguyen, P.V., & Abel, T. (2007). Genetic disruption of

    protein kinase A anchoring reveals a role for compartmentalized kinase signaling in theta-burst long-

    term potentiation and spatial memory. Journal of Neuroscience 27, 10278-10288.

    Nijholt, I.M., Ostroveanu, A., de Bruyn, M., Luiten, P.G., Eisel, U.L., & Van der Zee E.A. (2007).

    Both exposure to a novel context and associative learning induce an upregulation of AKAP150

    protein in mouse hippocampus. Neurobiology of Learning and Memory, 87, 693-696.

    Nijholt I., Ostroveanu A., Scheper W., Penke B., Luiten P., Van der Zee E., & Eisel U. (2008).

    Inhibition of PKA anchoring to A-kinase anchoring proteins impairs consolidation and facilitates

    extinction of contextual fear memories. Neurobiology of Learning and Memory 87, 693-696.

    Nijholt I.M., Dolga A.M., Ostroveanu A., Luiten P.G., Schmidt M., & Eisel U.L. (2008). Neuronal

    AKAP150 coordinates PKA and Epac mediated PKB/AKT phosphorylation. Cellular Signaling 10,

    1715-1724.

    Oestreich, E.A., Wang, H., Malik, S., Kaproth-Joslin, K.A., Blaxall, B.C., Kelley, G.G., Dirksen,

    R.T., & Smrcka, A.V. (2007). Epac-mediated activation of phospholipase C(epsilon) plays a critical

    role in beta-adrenergic receptor-dependent enhancement of Ca2+ mobilization in cardiac myocytes.

    Journal of Biological Chemistry 8, 5488-5495.

    Oh, M.C., Derkach, V.A., Guire, E.S., & Soderling, T.R. (2006). Extrasynaptic membrane trafficking

    regulated by GluR1 serine 845 phosphorylation primes AMPA receptors for long-term potentiation.

    Journal of Biological Chemistry 2, 752-758.

    Ostroveanu, A., Van der Zee, E.A., Dolga, A.M., Luiten, P.G., Eisel, U.L. & Nijholt, I.M. (2007). A-

    kinase anchoring protein 150 in the mouse brain is concentrated in areas involved in learning and

    memory. Brain Research 1145, 97-107.

    Ostroveanu, A., Van der Zee, E.A., Eisel, U.L. & Nijholt, I.M. (2009). Detailed analysis of mAKAP

    expression in the brain of young and old mice. Submitted

    Ostroveanu, A., Van der Zee, E.A., Eisel, U.L., Schmidt, M., & Nijholt, I.M. (2009). Exchange

    protein activated by cyclic AMP 2 (Epac2) plays a specific and time-limited role in memory retrieval.

    Submitted

    167

  • Chapter 7

    Ouyang, M., Zhang, L., Zhu, J.J., Schwede, F., & Thomas, S.A. (2008). Epac signaling is required for

    hippocampus-dependent memory retrieval. Proceedings of the National Academy of Sciences United

    States of America 105, 11993-11997.

    Pare, G.C., Bauman, A.L., McHenry, M., Michel, J.J., Dodge-Kafka, K.L., & Kapiloff, M.S. (2005).

    The mAKAP complex participates in the induction of cardiac myocyte hypertrophy by adrenergic

    receptor signaling. Journal of Cell Science 118, 5637-5646.

    Pereira, L., Métrich, M., Fernández-Velasco, M., Lucas, A., Leroy, J., Perrier, R., Morel, E.,

    Fischmeister, R., Richard, S., Bénitah, J.P., Lezoualc'h, F., & Gómez, A.M. (2007). The cAMP

    binding protein Epac modulates Ca2+ sparks by a Ca2+/calmodulin kinase signalling pathway in rat

    cardiac myocytes. The Journal of Physiology 2, 685-694.

    Pilcher, W.H., & Joseph, S.A. (1984). Co-localization of CRF-ir perikarya and ACTH-ir fibers in rat

    brain. Brain Research 299, 91-102.

    Rangarajan, S., Enserink, J.M., Kuiperij, H.B., de Rooij, J., Price, L.S., Schwede, F., & Bos, J.L.

    (2003). Cyclic AMP induces integrin-mediated cell adhesion through Epac and Rap1 upon

    stimulation of the beta 2-adrenergic receptor. The Journal of Cell Biology 4, 487-493.

    Rosenmund, C., Carr, D.W., Bergeson, S.E., Nilaver, G., Scott, J.D., & Westbrook, G.L., (1994).

    Anchoring of protein kinase A is required for modulation of AMPA/kainate receptors on hippocampal

    neurons. Nature 368, 853-856.

    Ruehr, M.L., Russell, M.A., Ferguson, D.G., Bhat, M., Ma, J., Damron, D.S., Scott, J.D., & Bond, M.

    (2003). Targeting of protein kinase A by muscle A kinase-anchoring protein (mAKAP) regulates

    phosphorylation and function of the skeletal muscle ryanodine receptor. Journal of Biological

    Chemistry 278, 24831-24836.

    Schwaerzel, M., Jaeckel, A., & Mueller, U. (2007). Signaling at A-kinase anchoring proteins

    organizes anesthesia-sensitive memory in Drosophila. Journal of Neuroscience, 27, 1229-1233.

    Shankar, G.M., Li, S., Mehta, T.H., Garcia-Munoz, A., Shepardson, N.E., Smith, I., Brett, F.M.,

    Farrell, M.A., Rowan, M.J., Lemere, C.A., Regan, C.M., Walsh, D.M., Sabatini, B.L., & Selkoe, D.J.

    (2008). Amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic

    plasticity and memory. Nature Medicine 8, 837-842.

    168

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Pereira%20L%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Fern%C3%A1ndez-Velasco%20M%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Lucas%20A%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Leroy%20J%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Morel%20E%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Fischmeister%20R%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Richard%20S%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22B%C3%A9nitah%20JP%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Lezoualc'h%20F%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22G%C3%B3mez%20AM%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/pubmed/12578910?ordinalpos=28&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/pubmed/12578910?ordinalpos=28&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Shankar%20GM%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Li%20S%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Mehta%20TH%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Garcia-Munoz%20A%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Shepardson%20NE%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Smith%20I%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Brett%20FM%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Farrell%20MA%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Lemere%20CA%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Regan%20CM%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Walsh%20DM%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Sabatini%20BL%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Selkoe%20DJ%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlus

  • General discussion

    Sik, A., Gulacsi, A., Lai, Y., Doyle, W.K., Pacia, S., Mody, I., & Freund, T.F., (2000). Localization

    of the A kinase anchoring protein AKAP79 in the human hippocampus. European Journal of

    Neuroscience 12, 1155-64.

    Sinnegger-Brauns, M., Hetzenauer, A., Huber, I., Renström, E., Wietzorrek, G., Berjukov ,S., Cavalli,

    M., Walter, D., Koschak, A., Waldschütz, R., Hering, S., Bova, S., Rorsman, P., Pongs, O.,

    Singewald, N., & Striessnig, J. (2004). Isoform-specific regulation of mood behavior and pancreatic

    βcell and cardiovascular function by L-type Ca 2+ channels. Journal of Clinical Investigation 10,

    1430-1439.

    Smith, K.E., Gibson, E.S., & Dell'Acqua, M.L. (2006). cAMP-dependent protein kinase postsynaptic

    localization regulated by NMDA receptor activation through translocation of an A-kinase anchoring

    protein scaffold protein. The Journal of Neuroscience 9, 2391-2402.

    Snyder, E.M., Colledge, M., Crozier, R.A., Chen, W.S., Scott, J.D., & Bear, M.F. (2005). Role for A

    kinase-anchoring proteins (AKAPS) in glutamate receptor trafficking and long term synaptic

    depression. Journal of Biological Chemistry 280, 16962-16968.

    Soderling, S.H., Langeberg, L.K., Soderling, J.A., Davee, S.M., Simerly, R., Raber, J., & Scott, J.D.

    (2003). Loss of WAVE-1 causes sensorimotor retardation and reduced learning and memory in mice.

    Proceedings of the National Academy of Sciences United States of America 100, 1723-1728.

    Ster, J., De Bock, F., Guérineau, N.C., Janossy, A., Barrère-Lemaire, S., Bos, J.L., Bockaert, J., &

    Fagni, L.(2007). Exchange protein activated by cAMP (Epac) mediates cAMP activation of p38

    MAPK and modulation of Ca2+-dependent K+ channels in cerebellar neurons. Proceedings of the

    National Academy of Sciences United States of America 104, 2519-2524.

    Ster, J., de Bock, F., Bertaso, F., Abitbol, K., Daniel, H., Bockaert, J., & Fagni, L. (2009). Epac

    mediates PACAP-dependent LTD in the hippocampus. The Journal of Physiology 1, 101-113.

    Tavalin, S.J., Colledge, M., Hell, J.W., Langeberg, L.K., Huganir, R.L., & Scott, J.D., (2002).

    Regulation of GluR1 by the A-kinase anchoring protein 79 (AKAP79) signaling complex shares

    properties with long-term depression. Journal of Neuroscience 22, 3044-3051

    Tunquist, B.J., Hoshi, N., Guire, E.S., Zhang, F., Mullendorff, K., Langeberg, L.K., Raber, J., &

    Scott, J.D. (2008). Loss of AKAP150 perturbs distinct neuronal processes in mice. Proceedings of the

    National Academy of Sciences United States of America 34, 12557-1262.

    169

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Smith%20KE%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=pubmed&Cmd=Search&Term=%22Gibson%20ES%22%5BAuthor%5D&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DiscoveryPanel.Pubmed_RVAbstractPlushttp://www.ncbi.nlm.nih.gov/pubmed/18711127?ordinalpos=1&itool=EntrezSystem2.PEntrez.Pubmed.Pubmed_ResultsPanel.Pubmed_DefaultReportPanel.Pubmed_RVDocSum

  • Chapter 7

    Wemmie, J.A., Chen, J., Askwith, C.C., Hruska-Hageman, A.M., Price, M.P., Nolan, B.C., Yoder,

    P.G., Lamani, E., Hoshi, T., Freeman, J.H. Jr, & Welsh, M.J. (2002).The acid-activated ion channel

    ASIC contributes to synaptic plasticity, learning, and memory. Neuron 3, 463-477.

    Wemmie, J.A., Askwith, C.C., Lamani, E., Cassell, M.D., Freeman, J.H. Jr, & Welsh, M.J. (2003).

    Acid-sensing ion channel 1 is localized in brain regions with high synaptic density and contributes to

    fear conditioning. Journal of Neuroscience 13, 5496-5502.

    Wemmie, J.A., Coryell, M.W., Askwith, C.C., Lamani, E., Leonard, A.S., Sigmund, C.D., & Welsh,

    M.J. (2004). Overexpression of acid-sensing ion channel 1a in transgenic mice increases acquired

    fear-related behavior. Proceedings of the National Academy of Sciences United States of America 10,

    3621-3626.

    Yao, J.-J., Huang, Z.-H., Masten, S.J., Mizutani, T., Nakashima, S., & Nozawa, Y. (1998). Changes in

    the expression of protein kinase C (PKC), phospholipases C (PLC) and D (PLD) isoforms in spleen,

    brain and kidney of the aged rat: RT-PCR and Western blot analysis Mechanisms of Ageing and

    Development 1, 151-172.

    Zhong, N., & Zucker, R.S. (2005). cAMP acts on exchange protein activated by cAMP/cAMP-

    regulated guanine nucleotide exchange protein to regulate transmitter release at the crayfish

    neuromuscular junction. Journal of Neuroscience 25, 208-214.

    170