Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Current Topics in Medicinal Chemistry, 2011, 11, 1393-1405 1393

The cAMP-Dependent Protein Kinase Pathway as Therapeutic Target –


Possibilities and Pitfalls

Rune Kleppe1,*, Camilla Krakstad2, Frode Selheim1,3, Reidun Kopperud1 and Stein Ove Døskeland1

1
Department of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway, 2Department of Clinical
Medicine, Haukeland University Hospital, 5021 Bergen, Norway, 3Proteomic Unit (PROBE), University of Bergen, Ber-
gen, Norway

Abstract: The prototype second messenger cAMP and its major mediator, the cAMP-dependent protein kinase (PKA), is
able to control simultaneously multiple processes within the same cell. This appears to be achieved through its unique dis-
sociative regulation and the spatiotemporal regulation of both cAMP and PKA. The widespread tissue distribution and
physiological function of this pathway makes it an attractive, but challenging pharmacological target. We will discuss cur-
rent progress in manipulating the fine-tuning of PKA, and outline so far underexploited possibilities for therapy, such as
novel ways to target specific substrates and catalytic cycle intermediates of PKA. An attractive strategy to achieve a more
focused pharmacological treatment is to combine more traditional targeting of extracellular receptors or ligands with that
of intracellular signaling pathway components. The cAMP signaling pathway provides a variety of possibilities for such
an approach.
Keywords: Cell signaling, PKA, cAMP, multi-targeting, disease.

1. INTRODUCTION tory diseases like COPD and asthma [5]. More recently, iso-
type selective inhibitors, such as the PDE5 inhibitor silde-
Intracellular signaling is instrumental in coordinating nafil (Viagra), and the PDE3 inhibitor milrinone have been
cellular responses to external cues, but the implication of
developed. Sildenafil has proven successful in treating erec-
aberrant signaling in disease is only recently becoming ap-
tile dysfunction [6], while short-term administration of mil-
preciated. The complexity of signaling and the relative recent
rinone increase vasodilation and cardiac contraction after
status of signaling components as drug targets suggest that
heart failure [7].
signaling modulation will be a rich source of potential drug
candidates for years. The cyclic nucleotides are prototype The limited distribution of PKG restricts the number of
second messengers whose major downstream mediators are diseases that can be targeted, but makes it also easier to
protein kinases. The cAMP-dependent protein kinase (PKA) achieve focused therapy with fewer side effects. In contrast,
is considered a protein kinase paradigm. The compact cata- the widespread tissue-distribution and broad functional rele-
lytic subunit is inhibited by a (pseudo)substrate sequence vance of cAMP signaling provides more opportunities, but
located on a separate cAMP binding regulatory subunit, also higher risk of side effects. A possible solution to the
rather than by an inhibitory domain in the same polypeptide dilemma of low specificity may be found in the great variety
chain [1]. Activation of PKA therefore involves subunit dis- of signaling proteins and signaling mechanisms involved in
sociation, which is unique among kinases, and differs controlling the cAMP pathway. The rich isotypic variation
strongly from the activation of its evolutionary offspring the combined with extensive spatiotemporal regulation and
cGMP-dependent protein kinase (PKG), whose cGMP bind- cross-signaling provides plentiful of opportunities to fine-
ing sites reside in the same polypeptide chain as the catalytic tune the selectivity. Some of these will be discussed here
site [2]. The activity of PKA and its cGMP-activated relative with emphasis on specific targeting of the PKA isozymes
are already being modulated pharmacologically through ex- and their downstream protein substrates.
tracellular receptor agonists/antagonists and by inhibitors of
cNMP degrading phosphodiesterases. At the receptor level, 2. THE cAMP SIGNALING SYSTEM - OVERVIEW
antagonists for the ß1-adrenergic receptors have been used in 2.1. Regulation of cNMP Turnover
treatment of angina and hypertension (e.g. metoprolol),
while the ß2-adrenergic receptor agonists (e.g. salmeterol) The second messenger cAMP is synthesized from ATP
are used for treating asthma or chronic obstructive pulmo- by adenylyl cyclases (ACs) (see Fig. (1) for overview),
nary disease (COPD) [3, 4]. Both general and more selective whereas cGMP is synthesized from GTP by guanylyl cy-
PDE-inhibitors have reached the clinic. The general PDE clases (GCs). The eukaryotic AC families belong to AC class
inhibitor theophylline is used for treatment of respira- III, and can be either soluble (sAC) or of the more com-
monly transmembrane (tmAC) type (reviewed in [8]). Nine
*Address correspondence to this author at the Department of Biomedicine,
genes (I-IX) code for mammalian tmAC, and each can exist
University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; in several splice variants. TmACs have low basal activity,
Tel: +47-555-86360; Fax: +47-555-86375; are activated by the heterotrimeric Gs, are often subjected
E-mail: rune.kleppe@biomed.uib.no to additional positive and negative modulation by other G-

1568-0266/11 $58.00+.00 © 2011 Bentham Science Publishers Ltd.


1394 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 Kleppe et al.

tmAC
GPCR GPCR
Ligand
Ligand

Gαs Gα12 Gβγ


PM Gβγ
ATP Epac
AMP
cAMP( ) C GTP- Rap1 Rap1 -GDP
P

RII RII
PDE AKAP-lbc GAP
P P Adhesion
C P
Secretion
C Novel targets
C RI RI PKAII
PDE
C PDE C
PKAI RI
R I
C RI
RI C
C

C C
C sA
C
PKI ATP
P
CREB

RI
cAMP( )

RI
C
ICER

Nucleus Activating
Cytosol Inhibiting
Translocation

Fig. (1). Overview of the cAMP signaling pathway. The Figure shows a cartoon of the cAMP pathway initiated by ligand binding to G-
protein coupled receptors (GPCR) and activation of heterotrimeric G-proteins. The dissociated GS activated transmembrane adenylyl cy-
clases (tmAC) to synthesize cAMP (grey dots, grey shaded color represents cAMP concentration gradients), which is degraded by phosphodi-
esterases (PDEs). Downstream targets of cAMP are PKA type I and II and Epac. The dimeric regulatory subunits of PKA-I (RI) and (RII) are
shown with empty or filled cAMP-binding sites. Binding of two cAMP molecules/R-subunit enables dissociation of the catalytic subunit,
which may then phosphorylate downstream targets in different compartments within the cell. Anchored PKA-II is shown bound to AKAP-
lbc. Nuclear proteins are cAMP-response element binding protein (CREB), PKA-inhibitor (PKI) and immediate cAMP-induced early re-
pressor (ICER). Soluble AC is shown as sAC.

protein subtypes (see also [9] for review). ACI and ACVIII the more than 50 enzyme variants span a range of enzyme
are activated by Ca2+/calmodulin [10], and several AC iso- kinetic types with different Km, cNMP specificity, and regu-
types are regulated by phosphorylation. One gene is identi- lation by other second messengers such as calcium. For the
fied for sAC. This ubiquitously expressed enzyme is stimu- dual nucleotide PDEs the cNMPs are mutual competitive
lated by bicarbonate, calcium and lactate [8, 11]. For this inhibitors, depending on their relative affinities and Vmax
reason the sACs have been suggested to represent an evolu- values. A special example of this is PDE3A, which has high
tionary conserved family of proteins involved in sensing the affinities for both cAMP and cGMP, and is referred to as a
cellular metabolic state [12]. cGMP-inhibited cAMP-PDE due to a 10 fold higher V max for
cAMP compared to cGMP [16]. Several of the PDE enzyme
The level of free cAMP and cGMP (cNMP) in cells is
controlled by the opposing action of AC/GC and PDEs Fig. types have cGMP binding GAF domains (mammalian
cGMP-binding PDEs, Anabaena Adenylyl cyclases, and
(1). The mammalian PDE superfamily so far constitutes 11
Escherichia coli FhlA), which mediate allosteric activation
families, each of which originates from 1-4 genes, with sev-
of the catalytic domain [17]. In addition to the different ki-
eral isoforms (see [13,14,15] for recent reviews). Altogether,
netic characteristics, several splice variants locate to differ-
The cAMP-Dependent Protein Kinase Pathway as Therapeutic Target Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 1395

ent cell compartments, through membrane association and early embryonic development [41]. The C-/- knockout
binding to proteins, such as to A-Kinase anchoring proteins mouse is viable, but has a severely growth retarded pheno-
(AKAPs) in the case of the PDE4 family [18]. The fact that type and approximately 73 % of the animals die before
so many gene products are devoted to the synthesis and deg- adulthood [35]. In contrast, the C-/- mouse has no apparent
radation of cyclic nucleotides indicates that the regulation of alteration of phenotype [42].
the cNMP concentration is of high priority. The multitude of
The C seems to be specific to human testis [29]. PrKX
splice variants, post-translational modifications and protein
(X chromosome-encoded protein kinase X) differ from the
interaction partners obviously provides cells with rich oppor-
other C-subunits by selectivity binding to the RI subunits
tunity to control the dynamics of cNMPs and to couple
and by phosphorylation of the RII subunits in a cAMP inde-
cAMP and cGMP signaling together as well as to other sig-
pendent manner. PrKX is expressed in several tissues, with
naling pathways. highest mRNA level detected in fetal and adult brain, kidney
and lung [43].
2.2. Receptors for cAMP – PKA
In its inactive states, PKA is a holoenzyme, consisting of 2.3. Receptors for cAMP – Epac and cAMP-gated Ion
a dimeric regulatory moiety, composed of two regulatory (R) Channels
subunits, and up to two catalytic (C) subunits. The C-
Epac proteins function as guanine nucleotide exchange
subunit(s) becomes catalytically competent only upon disso-
factors for both Rap1 and Rap2, which belong to the Ras
ciation from the regulatory subunits, which is facilitated by family of small GTP-binding protein [44] (Fig. (1)). Two
binding of cAMP to the R-subunit to which it is associated
isoforms have been identified, Epac1 and Epac2, which are
(see Fig. (1), text below). The N-terminal of the R subunit
encoded by two different genes [45]. Recently, three splice
contains a dimerization domain, and a localization domain
variants of Epac2 were identified and named Epac2A, B and
responsible for binding the R dimer to A-kinase anchor pro-
C [46,47]. Epac1 and Epac2 share the same functional do-
teins (AKAPs). The N-terminal domains of R are followed
mains with a N-terminal regulatory region that contains one
by an autophosphorylation/autoinhibitory motif, and two (Epac1, Epac2B, Epac2C) or two (Epac2A) cyclic nucleotide
homologous cAMP binding sites, referred to as A- and B-
binding-domains and a Dishewelled/Egl-10/Pleckstrin do-
site, at the C-terminal [19,20]. The cAMP binding sites have
main (Epac1, Epac2A, Epac2B). The catalytic region is at
different kinetic properties and both sites must be occupied
the C-terminal and contains a CDC25 homology domain for
to achieve activation [21].
GTP-exchange activity in addition to a Ras exchange motif
Two mammalian PKA isotypes (PKA-I and II) have been domain and a Ras association domain [44]. Upon binding of
identified, composed by dimeric type I or type II R subunits. cAMP, a conformational change take place and the active,
Each isotype of R can be further subdivided into  and  catalytic region is exposed [48,49]. This conformational
isoforms, encoded by different genes [22]. The different iso- change has also been linked to subcellular localization of
forms of R-isotypes show somewhat different tissue expres- Epac1, e.g. to the plasma membrane [50].
sion. In general, the  isoform of RI and RII are ubiquitously
The Epac proteins are expressed at different levels in
expressed, whereas the  isoforms are mainly expressed in most tissues. While Epac1 is expressed ubiquitously, recent
nervous system, lung and gonads [23]. Knock-out of RI in
studies indicate that the expression of Epac2 splice variants
mouse results in early embryonic death [24], suggesting that
is tissue specific. Epac2A dominates in pituitary glands and
this isoform play a key role in the PKA-signaling system.
pancreatic -cells, while Epac2B is the main variant in adre-
Consistent with that notion, this isoform was found to be
nal glands. Epac2B is expressed in adrenocortical cells and is
regulated in compensation to genetic deletion of R isoforms
involved in cAMP-induced actin reorganization and cell mi-
and during overexpression of the catalytic subunit gration together with PKA [51]. The third variant, Epac2C,
[23,24,25]. Deletion of the neuronal RI isoform, RI, show
is specifically expressed in the liver [46,47].
reduced short-term memory, despite compensatory expres-
sion by RI and invariant total PKA activity [25]. Unlike for Ion channels regulated by cAMP are part of the cyclic
RI, mice lacking RII appear healthy, whereas the RII nucleotide-gated ion channel family. They are nonselective
knock-out show protection against diet-induced obesity cation channels that allow the passage of K+, Na+ and Ca2+
[25,26]. (see [52] for review), and their activation represents an im-
portant step in the signal transduction pathway in both vision
Four genes are known for the mammalian C subunit;
(cGMP) and olfaction (cAMP and cGMP) [53]. Cyclic nu-
C [27], C [28], C [29,30] and PrKX [31]. C is further
cleotide-gated ion channels have also been found in a variety
divided into three different forms caused by post-
of other cell types [54,55].
translational modification (C1 and C2) and alternative
splicing (C-s) [32,33,34]. The C1 is ubiquitously ex-
pressed while C-s is found in sperm, where it is involved in 2.4. Activation of PKA – a Unique Mechanism
mobility [35]. The C gene transcribes several splice vari- Dissociative activation makes PKA unique within the
ants; C1, C2, C3 and C4 [36] of which C1 is ubiqui- mammalian kinome. This feature has been kept throughout
tously expressed [37], C2 is predominantly expressed in evolution from yeast even though it has been lost in evolu-
lymphoid cells [38,39], and C3 and C4 variants are found tionary related proteins such as the cGMP-dependent kinase.
solely in neuronal tissues [40]. The difference between these Separated regulatory (R) and catalytic (C) protomers enables
splice variants is in the N-terminal region of the protein [38]. different possibilities for regulation than for single chained
Genetic null mutations of both C and C result in death at kinases. A consequence of separated catalytic and regulatory
1396 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 Kleppe et al.

chains, is that the degree of dissociation is controlled by cally recognizes the far N-terminal region of the regulatory
mass action kinetics. Thus, the degree of activation depends subunits, which form a docking domain in the immediate
on the concentration of kinase in cellulo and the dissociation spatial vicinity of the dimerization motifs of the regulatory
constant between the catalytic subunit and the cAMP- subunits [71,72,73]. Presently, about 50 different A-kinase
saturated regulatory subunit. It is generally believed that anchoring proteins (AKAPs) have been described (including
PKA-I is half activated between 50-100 nM cAMP and that splice variants) (see [74] for recent review), the majority of
full saturation of the holoenzyme leads to complete dissocia- which bind PKA-II. However, PKA-I specific AKAPs have
tion and activation [56, 57]. However, more recent findings been reported also, such as the 4 integrin receptor subunit
suggest that this is not the case. In fact the dissociation con- [75], and dual specificity AKAPs that are capable of binding
stants for ubiquitous PKA-I and II isotypes are lower than both PKA isotypes [76]. The battery of different AKAPs
previously thought and in a range that sensitizes the dissocia- therefore present a wealth of possibility for cells to localize
tive activation to the expression levels of the regulatory PKA isotypes to the cytosolic face of almost any organelle or
subunits [58,59]. The thermodynamic driving force for the cellular structure. Many of the AKAPs also bind to other
activation is a 1000 fold higher affinity for cAMP to free signaling proteins, such as PDEs (PDE4 isoforms), protein
regulatory subunit relative to the holoenzyme [60]. Still it phosphatases and other kinases. In particular the co-
seems that cAMP is not capable of fully activating PKA, at localization with PDE and recently also AC, is thought to be
least in cells that express high levels of kinase. In this re- a crucial feature of spatially confined cAMP-signaling do-
spect, the possible existence of super-activators is particular mains [77, 78].
interesting.
Several issues are however unclear regarding PKA com-
Recent progress in the structural understanding of PKA partments, such as the local concentration of the active com-
activation has come from comparison of cAMP-bound R- ponents in cAMP signaling or the diffusion flux of these
subunit and the resolved structures of PKAI and II holoen- proteins and cAMP itself in and out of these compartments.
zymes [1,61,62,63,64,65]. The structures of the holoenzymes Localized PDE has been shown to control cAMP leakage out
show large surface interactions between the R and C of compartments (see for [79] review) in addition to regulat-
subunits which are greatly diminished upon binding of ing the local cAMP level and duration of the cAMP signal.
cAMP (reviewed in [66]). Resolving the interface between C Selective inhibition of PDE is therefore one approach to in-
subunit and cAMP-saturated R subunit at the structural level crease the compartment signal, but might generate signifi-
is however a major challenge that is probably only possible cant spillover of cAMP and activate cAMP-receptors outside
through high-field NMR due to fast on-/off- interactions of the borders of compartment if not properly dosed. For PDEs
the subunits. Possibly, the use of single cAMP-site mutants that are controlled by additional allosteric modulators such
of the R-subunit can resolve some of the structural enigmas as cGMP and Ca2+/calmodulin, selective pharmacotargeting
of PKA activation by cAMP binding, as the affinity between offers a possibility to modulate the cross-signaling between
cAMP-bound R and C-subunit is still in the nanomolar cAMP and other signaling pathways within the compart-
range. Recent NMR studies have revealed some of the struc- ment.
tural mechanisms underlying communication between the
The responsiveness of a compartment to a signal is how-
cAMP-binding sites of the RI subunit [62,65].
ever not only determined by the cAMP dynamics. The PKA
Mass action controlled activation enables additional lay- activation dynamics and the protein phosphatase activity are
ers of regulation by controlling the level of R- and C- also important determinants. These issues are much less re-
subunits. Thus, different R to C stoichiometries as well as solved than for cAMP, in particular the well known fact that
feedback and feedforward regulatory mechanisms can be the catalytic subunit can diffuse freely when activated. The
envisioned, some of which have been found experimentally repetitive signaling capacity of compartments might there-
(see [67] for review). Separated catalytic and regulatory fore be limited by their ability to contain or reload with cata-
subunits will also influence the use of PKA for compartmen- lytic subunit. The observation that the R-subunit and the dif-
talized signaling as diffusion will act to separate concentra- fusing catalytic subunit form complexes, even at high levels
tion differences within the cell. In that respect, single chain of cAMP, may provide answers to some of these questions.
kinases such as PKG would be expected to be better suited In particular to address the different characteristics of com-
for kinase activation in signaling domains that are spatially partments containing type I versus type II kinase.
restrained. Still, spatial regulation of PKA seems to be more
widespread than for many of the evolutionary related 3. THE PHYSIOLOGY AND PATHOPHYSIOLOGY
kinases. OF cAMP SIGNALING
3.1. Metabolism, Energy Expenditure and Deposition
2.5. Spatially Confined Signaling Through PKA
Localization of PKA to cellular structures was described PKA was the first discovered effector of cAMP and was
first implicated in regulation of glycogen metabolism [80].
in the early seventies in brain tissue where a majority of
Thus, regulation of energy metabolism and blood glucose
PKA is associated with non-soluble fractions [68]. However,
levels were the first described functions of PKA. Perhaps not
the ability of proteins to physically bind PKA was first de-
surprisingly, a function in regulation of energy store usage
scribed for the microtubule associated protein 2 [69] and
was found for PKA rather simultaneously in tissues such as
found to be mediated through an A-kinase anchoring do-
main, an amphipathic helix motif first described in Ht31/ skeletal muscle, heart, liver and adipose tissue. More re-
cently, the concerted action of the cAMP effectors PKA and
AKAP-Lbc/AKAP13 [70]. The anchoring domain specifi-
Epac was described in differentiation of white adipose tissue
The cAMP-Dependent Protein Kinase Pathway as Therapeutic Target Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 1397

[81]. Thus, it seems that cAMP signaling is linked to both multitude of functions have been ascribed to this kinase in
fatty acid usage and increase of white adipose tissue. Studies the CNS. There is now clear evidence that PKA signaling
on knock-out mice of PKA and AC3 supports a role for has a key role in a variety of implicit and explicit memory
cAMP-signaling through PKA in the regulation of energy processes in both invertebrate and mammals [99,100]. Re-
usage and appetite [82,83,84,85], and this pathway may cently, a transgenic mouse expressing a dominant-negative
therefore play an important role in the development of meta- form of the PKA regulatory subunit (RI) in two regions of
bolic syndrome and obesity. hippocampus, was found to have a deficit in several forms of
long-term synaptic potentiation (LTP) and a lowered thresh-
3.2. Cardiovascular System, Blood Cells old for the consolidation of long-term synaptic depression
(LTD) [101]. PKA is crucial for the dynamic balance be-
In the cardiovascular system, cAMP and cGMP play im- tween phosphatases and protein kinases, which regulate the
portant roles in most cell types. Increased cardiac perform- consolidation of LTP and LTD. A dominant negative RI
ance is a well described function of cAMP and PKA through results in decreased kinase activity and thereby less phos-
adrenergic amplification of cardiomyocyte calcium signal- phorylation of the AMPA glutamate receptor, leading to sus-
ing. Together with Epac, PKA is also implicated in - tained LTD [102].
adrenergic stimulated hypertrophy [86], but this is yet to be
confirmed in transgenic animal models. For vascular endo- Serotonergic and catecholaminergic neurotransmission
thelial cells, stabilization of the barrier function of the endo- are implicated in the modulation of gross neurological func-
thelial cell layer in response to permeabilizing agents (such tions such as mood, sleep, memory, motivation, pleasure and
as LPS, thrombin, TNF- and TGF-) is the best described motor function. The neurotransmission of these monoamines
effect of cAMP [87,88,89]. This is reported to occur through is mediated through a range of receptors of which several are
both Epacs and PKA, where modulation of the activities of linked to cAMP signaling [103,104]. In addition, PKA is
the Rho-GTPases is a major consensus mechanism [89, 90, implicated in the regulation of biosynthesis of catechola-
91, 92]. Intriguingly, cAMP has been implicated in adhesion mines and serotonin [105,106]. Not surprisingly, cAMP-
and chemotaxis of leukocytes, as well as in the inhibition of signaling has been suggested as a potential target in mood
smooth muscle proliferation and constriction, which together disorders and Parkinson’s disease [107,108]. This is also
with its beneficial effect on endothelial barrier function, have supported by examination of PKA protein levels in human
made cAMP-signaling a possible target in atherosclerosis post-mortem brain tissue of normal and depressed persons
and pulmonary hypertension. The PDE4 inhibitor roflumilast [109,110]. In patients with the neurodegenerative Alz-
has shown promising potential in several preclinical trials for heimer’s disease, the level of PKA was reduced, resulting in
treatment of COPD [93]. deficient CREB function. This down regulation was due to
degradation of the PKA subunits and may result from over-
In platelets, cyclic nucleotide signaling through cAMP activated calpain [111].
and cGMP are important antagonistic pathways that inhibits
activation and aggregation of platelets in response to agents
3.4. Immune System, Pathogenic Bacteria
such as thrombin, collagen, von Willebrand Factor and ADP.
These inhibitory pathways are necessary to control the for- The cAMP-pathway is a well described repressor of
mation of thrombi (see [94] for review), which may result in macrophage activation and can therefore suppress inflamma-
stroke. Anti-platelet therapy is therefore used for secondary tory responses (reviewed in [112]). Much of this effect is
prevention of stroke, typically using acetylsalicylic acid (cy- PKA-mediated through inhibition of Toll-like-receptors,
clo-oxygenase inhibitor) or clopidogrel (ADP-receptor in- hence inhibiting the synthesis of inflammatory cytokines
hibitor) [95]. However, as these agents modify their target (e.g. TNF, macrophage inflammatory protein-1), but also
irreversibly, there is a risk of excessive bleeding. The benefit through both PKA and Epac1 by inhibition of phagocytic
of cNMPs in platelets is that they reversibly inhibit all the receptors and production of reactive oxygen species [113,
various platelet activation pathways, in response to vascular 114]. Not surprisingly, the repressing action of cAMP is hi-
endothelial stress. In platelets, cNMP levels are controlled by jacked by several pathogenic bacteria, most notably Myco-
PDE2, 3 and 5, whereas prostaglandins, adenosine and nitric bacterium tuberculosis. They grow within the phagosome of
oxide stimulates the synthesis of cAMP and cGMP, respec- the macrophages, thereby causing disease. Interestingly, it is
tively [96,97]. This set of PDEs provides interesting possi- found that an increased level of cAMP by bacterial AC,
bilities of cross-talk between the two pathways – positive stimulated growth of bacteria and virulence through host
through cGMP-inhibited PDE3 and negative through cGMP- PKA. The opposite situation was found at low cAMP levels,
activated PDE2. In particular PKA activation through PDE3 resulting in bacterial destruction [115].
is implicated in NO mediated inhibition of platelet shape
An immunomodulatory function of PKA is also found in
change, which is the initial reversible step in platelet activa-
T-cells, where localized PKA inhibits T-cell receptor activa-
tion [98]. Arguably, platelet shape change is an interesting tion through C-terminal Src Kinase (see [116] for review).
target for long term anti-platelet therapy due to its reversibil-
The cAMP/PKA pathway is also implicated in T-cell dys-
ity and upstream placement in platelet activation.
function. Acquired immunodeficiency syndrome results from
the dramatic loss of CD4+T-lymphocytes following human
3.3. Nervous System, Memory, Neurodegereration, Mood immunodeficiency virus type 1 (HIV-1) infection [117]. Re-
Disorders cently, PKA was found to phosphorylate and activate the
Apart from platelets, the central nervous system (CNS) HIV-1 viral protein R, which contributes to the CD4+ cell
contains the highest amount of PKA. Not surprisingly, a death [118]. Blocking PKA activity or the phosphorylation
1398 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 Kleppe et al.

of HIV-1 viral protein R may thereby represent a strategy for and poor activators of Epac [126]. Analogs with hydropho-
therapy against HIV-1 infection. In addition, it has been re- bic N6-substituents at the adenine ring of cAMP appear to
ported that elevated cAMP leads to a PKA-dependent in- bind site selective to the A-sites of RI and RII [127]. Thus,
crease in viral replication [119]. N6-benzoyl-cAMP, which preferentially bind the A-sites of
both PKA-I and PKA-II can be combined with BII-site bind-
4. STRATEGIES TO TARGET PKA-SIGNALING ing analogs, like Sp-5,6-DCl-cBIMPS or 8-piperidino-
cAMP, for selective activation of PKA-II. However, combi-
4.1. Targeting cAMP Levels nation of 8-piperidino-cAMP (AI- and BII-site selective)
Manipulation of second messenger levels by activation of with BI-site preferring analogs like 8-AHA-cAMP will lead
AC (e.g. forskolin, from the plant Coleus forskohlii) or inhi- to selective synergistic activation of PKA-I (Table 1). By
bition of PDEs (3-isobutyl-1-methylxanthine – IBMX) has using analogs such as the strong BI-site selective 8-AHA-
been a common strategy to activate PKA experimentally. cAMP, it was successfully demonstrated that the PKA-
This will however nonspecifically activate all downstream holoenzyme associated RI subunit bound cAMP with posi-
targets of cAMP, but has the potential of gaining specificity tive cooperativity through an interchain site A-B interaction
by targeting particular isoforms within the AC and PDE en- mechanism [60]. As mentioned above, selective synergistic
zyme families. For at least two decades, there has been an activation of PKA-I, PKA-II or both can be achieved by us-
interest in finding isoform specific inhibitors of the different ing different pairs of site selective cAMP analogs [67]. Such
PDEs, and this has led to several compounds with high se- synergy between pairs of analogs provides strong evidence
lectivity. Inhibition of PDE will unequally activate cyclic that the biological effect is mediated through PKA-signaling.
nucleotide signaling, however, targeting several of the PDEs Highly Epac-selective cAMP analogs possess a ribose 2'-
also has the potential of inhibiting cAMP (and cGMP) sig- O-methyl substituent, which impairs binding to PKA, and an
naling. Several isoforms of the PDE family have allosteric 8-substituent at the adenine ring of cAMP. 8-pCPT-2'-O-Me-
activator domains, GAF-domains, which upon ligand occu- cAMP is a strong Epac activator (Ka = 1.9 M vs Ka = 45
pation can increase the catalytic activity of the enzyme. The M for cAMP) that has more than 500 fold higher affinity
cGMP-activated PDE2 and 5 isoforms are examples of such. for Epac1 than for the A-site of PKA-I (Table 1)
The PDE2 isoforms conduct negative cross-talk between [81,126,128]. The Epac-activator synergized strongly with
cGMP and cAMP in several tissues such as the nervous sys- both the PKA-activator and NGF to promote adipogenesis
tem, the heart, platelets and vascular endothelium [120,121]. and PC-12 differentiation, as determined by neurite out-
Thus, in addition to PDE activation, specific antagonists to- growth [126,128]. Later it was shown that Epac activation
wards the GAF-domain of PDE2 would enable direct inhibi- per se induced differentiation of human SH-SY5Y neuro-
tion of this cross communication, without interfering with blastoma cells, an effect that was blocked by Epac1 ShRNA
the basal activity of the enzyme. Recently, GAF-ligands to- expression [129]. Importantly, recent work has implicated
wards PDE2 and 5 have been described [122]. This extra that 8-pCPT-2'-O-Me-cAMP can be hydrolyzed to bioactive
cNMP-binding domain provides exciting possibilities for metabolites with anti-proliferative effects in certain cell
pharmacological intervention. types [130]. In long term studies such undesirable side ef-
More recently, the use of forskolin derivatives have been fects can be avoided by using the Sp-modified and hydroly-
used to generate selectivity in the activation of different iso- sis-resistant form of 8-pCPT-2'-O-Me-cAMP. Unfortunately,
forms of AC (see [123] for review). The AC5 selective by using isothermal microcalorimetry, it is evident that Sp-8-
forskolin derivative, colforsin daropate hydrochloride, is pCPT-2'-O-Me-cAMPS can inhibit some cGMP- and cAMP-
now entering clinical use for advanced congestive heart fail- specific PDEs [131]. Moreover, the hydrolysis-resistant BII-
ure [124]. Generation of isoform specific inhibitors of ACs selective Sp-5,6-DCl-cBIMPS showed inhibitory effect
has proven challenging, due to the high conservation of the against some PDEs, especially PDE1B. In intact cells, ana-
active site between the isoforms. However, the inhibitor log-mediated inhibition of PDEs would lead to an increase in
KH7 shows selectivity towards sAC as compared to tmAC the intracellular levels of cyclic nucleotides. Thus, to avoid
[125]. possible misinterpretation of data, it is important to perform
cAMP studies with a variety of analogs. Moreover, the use
4.2. Targeting the cAMP Binding Sites of inhibitory Rp-modified cyclic nucleotide analogs, like Rp-
cAMPS and Rp-8-Br-cAMPS, should have effects opposite
The cyclic nucleotide activated kinases have been in the to the PKA analogs, but should not affect the action of Epac
forefront of developing specific kinase activators, through analogs.
chemical modifications of the cyclic nucleotide. The discov-
ery of Epac revealed that commonly used cAMP analogs like 4.3. Targeting the Catalytic Subunit
8-Br-cAMP and 8-pCPT-cAMP activated both PKA and
Epac. In recent years, cAMP-analogs have been developed Kinase inhibitors are often based on the ATP site of the
which discriminate well between Epacs and PKAs and even catalytic domain/subunit, and act as competitive inhibitors
between different isotypes of the PKAs. In Table 1 we sug- for this substrate, so called type I inhibitors (not to be con-
gest cAMP-analogs that can be used to obtain selective acti- fused with type I PKA). More recently, allosteric inhibitors
vation of cAMP receptors, which in addition are quite well of type II have been described, which stabilizes the inactive
characterized in terms of stability and interference with PDE kinase conformation [132]. A successful example of this
activities. The 6'-modified cAMP analogs N6-benzoyl-cAMP inhibitor type is the tyrosine kinase inhibitor imatinib used to
and N6-monobutyryl-cAMP are selective activators of PKA, target Bcr-Abl in Chronic myelogenous leukemia (see [133]
for review). However, a valuable lesson has been learned
The cAMP-Dependent Protein Kinase Pathway as Therapeutic Target Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 1399

Table 1. Relative affinities of selected cAMP-analogs to cAMP-binding sites. The table lists the affinity of analogs relative to cAMP
(Kd' = Kd,cAMP/ Kd, analog). Analog pair selectivity = (Kd' AI* Kd'BI)1/2 : (Kd' AII* Kd'BII)1/2. The data are from [60, 126]

RI (Kd', M) RII (Kd', M) Epac1 Analog Pair Selectivity
Analog
Site A Site B Site A Site B Kd' (M) RI/RII

N6-Bnz-cAMP 4.0 0.26 3.8 0.037 1.3


1:10
Sp-5,6-DCL-cBIMPS 0.022 0.13 0.034 14 0.69

8-Pip-cAMP 2.1 0.06 0.047 2.7 0.21


18:1
8-AHA-cAMP 0.055 4.1 0.010 0.39 1.3

8-pCPT-2'-O-Me-cAMP 0.043 0.0016 0.00059 0.024 4.6

about the price to pay for high specificity, which is higher ating PKA from its anchoring site. Perhaps the most chal-
risk of resistance through missense mutations [133]. This is a lenging point of this strategy is to find compounds with both
particular risk with cancer drugs where genetic instability disruptive and pharmacological potential, due to a large hy-
compared with high selection pressure drive the disease pro- drophobic interphase between AKAPs and PKA. In addition,
gression and its response to therapy. Other diseases are gen- specificity can be difficult to achieve due to high similarity
erally less prone to such problems, except through natural at the anchoring domain of most AKAPs, particularly be-
polymorphisms within the population. tween the PKAII specific AKAPs. However, a PKA isotype
directed disruption seems feasible in light of recent progress
For the PKAs, no ATP-site competitive inhibitors are to
on RI-anchoring disruptor peptides, and may have promising
our knowledge available commercially, which display satis-
potential for treating certain T-cell dysfunctions [137,138].
factory selectivity. The ATP-competitive kinase inhibitors
H89 (of the isoquinolinesulfonyl group) and KT5720, which
are often used as PKA-specific inhibitors of the catalytic 5. NEW POSSIBILITIES - SUBSTRATE DIRECTED
subunits, have been shown to inhibit other kinases more CONTROL OF SIGNALING INVOLVING PROTEIN
potently than PKA [134]. H89 inhibits S6Kinase-1, mitogen- PHOSPHORYLATION
and stress-activated protein kinase 1, and Rho dependent 5.1. Modulation of Protein Phosphorylation Downstream
protein kinase-II (ROCK-II) with similar and higher potency of PKA
[134]. Thus, when investigating the influence of PKA on
processes that are controlled by Rho kinase, the use of H89 The highest specificity of pharmacological intervention
may be particularly misleading as H89 would inhibit PKA, of phosphorylation events is through manipulation at the
but still inhibit the ROCK, and RhoA inhibition is one the level of discrete kinase substrates. This can to some extent
established functions of PKA (see Fig. (2)). be achieved by modulating the kinase or phosphatase activi-
ties, but this will only provide specificity in the rare cases
Compared to the inhibitory pseudosubstrate region on the when the targeted kinase phosphorylates only very few sub-
regulatory subunits of PKA, the natural PKA heat stable in- strates. This is truly not the case for PKA. However, for
hibitors, PKI type I and II, inhibits the catalytic subunit by a many protein substrates their phosphorylated state(s) is again
similar and highly specific mechanism. The amino acid se- affected by association to other proteins, like the 14-3-3 fam-
quence of PKI containing the inhibition motif has been used ily members. 14-3-3 bind the phosphorylated target protein
to generate kinase inhibitors with high selectivity towards and thereby may change its activity, localization or interac-
PKA, e.g. PKI-14-22 (amino acids 14-22, myrostoylated for tion with other proteins (see [139] for review). Many binding
increased permeability). These peptide-based inhibitors are partners are described for the 14-3-3 proteins, amongst other
competitive, however, for kinases in the MAPK pathways, the related Rho-GEF proteins AKAP-Lbc and Lfc, which
inhibitor have been developed that are non-competitive to both bind 14-3-3 upon phosphorylation by PKA (see Fig.
both ATP and protein substrate (reviewed in [135]). A recent (2)) [140,141]. Binding of 14-3-3 to PKA-phosphorylated
strategy based on staurosporine has provided non- Rho-GEF inhibits its exchange activity, thereby mediating a
competitive PKA inhibitors with increased selectivity [136]. negative signaling connection between cAMP- and Rho-
However, as with present inhibitors targeting the catalytic signaling Fig. (2A and B). Therefore, as long as 14-3-3 is
subunit, there is to our knowledge no selectivity towards the bound, little Rho activation can occur through this Rho-GEF.
different isoforms of the C-subunit, and isotype selectivity
will be difficult to attain. This would potentiate the risk of In addition to its primary effect, binding of 14-3-3 inhib-
severe side-effects with systemic delivery, however, spatially its dephosphorylation and thereby prolongs the signal. The
controlled or cell-type directed delivery are alternatives to strength of the inhibition is expected to depend on the affin-
cope with such problems. ity between 14-3-3 and the phospho-protein. This affinity
can be modulated by substances that disrupt or enforce the
4.4. Targeting Localization interaction between 14-3-3 and its phospho-target. Since the
Disruption of localized PKA signaling can be an attrac- protein interaction interface is unique to each protein com-
tive strategy to obtain focused negative targeting, by dissoci- plex, it provides an opportunity to specifically target kinase
1400 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 Kleppe et al.

substrates. This strategy is illustrated in Fig. (2C) for the sible. Given that dissociation of Mg/ADP is rate limiting for
AKAP-Lbc, where a drug that act on the association strength the catalysis of PKA it is puzzling that even small peptide
between 14-3-3 and PKA-phosphorylated AKAP-Lbc (in substrates are phosphorylated with different Vmax values un-
Fig. (2C) to enforce the association) will act to modulate the less the ones with slow turnover stabilize the rate limiting
strength of the negative coupling between the cAMP and Mg/ADP-bound state of the kinase. This requires that sub-
Rho signaling pathways. Interestingly, the engineering of a strate binds to Mg/ADP-bound C-subunit, an option com-
compound that stabilize the interaction between 14-3-3 and a monly not considered in schemes describing protein kinase
binding partner is used by the fungus Fusicoccum amygdali, kinetics. We have evidence that the standard peptide sub-
which produces the wilt-inducing phytotoxin fusicoccin. The strate kemptide binds to Mg/ADP-bound C-subunit with
molecular mechanism for this toxin seems to be activation of similar affinity as to Mg/ATP-bound C-subunit, making it a
the plant plasma membrane H+-ATPase by stabilizing its realistic option [59] Fig. (3A,B). Expectedly, high affinity
interaction with 14-3-3 [142]. substrates would preferentially associate with the Mg/ADP-
bound form of the C-subunit (right reaction path Fig. (3)),
5.2. New Options to Exploit the Catalytic Mechanism of whereas low affinity substrates are expected to bind the C-
Phospho-Transfer for Kinase Activity Modulation subunit after nucleotide exchange to Mg/ATP (left reaction
path Fig. (3)). Compounds that accelerate ADP to ATP ex-
The catalytic mechanism of PKA has been well de- change would therefore be expected to discriminate in their
scribed. The phosphorylation appears to obey an ordered activation between these two substrate classes, depending if
kinetic mechanism, with Mg/ATP binding occurring first they facilitate exchange of the peptide-substrate free or
[143]. Still there does not seem to be any mutual interference bound form of the C-subunit (Fig. (3B)). It is however not
between Mg/ATP and peptide substrate binding, at least for currently known to what degree different PKA substrates
small peptides [144]. After formation of the ternary kinase- associate with the C-subunit in its different ligand states, and
Mg/ATP-peptide complex, the phospho-transfer occurs rap- to what degree their association affects nucleotide exchange.
idly, followed by an even faster phospho-substrate release (at
least for the model heptapeptide substrate Kemptide) [145]. The catalytic mechanism for different substrates becomes
Interestingly, the slow dissociation of ADP from the C- relevant during development of ATP-selective inhibitors,
subunit is rate limiting for the steady state kinase activity which may bind poorly to protein substrate associated forms
[145,146], and the Mg/ADP-bound form of the C-subunit of the C-subunit. Ligand free C-subunit only exists very
will be the dominating C subunit species when the kinase is transiently in cells, making it a poor target for inhibitors. On
active. The reaction scheme for PKA mediated phosphoryla- the other hand, the Mg/ADP-bound form is the most abun-
tion is shown in Fig. (3A). dant peptide-substrate-free form of the C-subunit and this
provides an opportunity to make compounds that trap the C-
Similarly to G-proteins where GDP to GTP exchange is subunit in a catalytically inactive state (I2 in Fig. (3B)). This
stimulatory, the catalytic efficiency of PKA could probably type of inhibitors would be expected to inhibit both high
be modulated by changing the exchange rate of ADP to affinity and low affinity substrates equally well. On the other
ATP. Binding of Mg2+ to a low affinity site, distinct from the hand, inhibitors that trap the Mg/ATP bound C-subunit
Mg/ATP binding site, slows down the dissociation of ADP (without substrate) in an inactive state are expected to pref-
considerably [146], indicating that this type of control is pos- erentially inhibit low affinity substrates (I1 in Fig. (3B)).

A Low cAMP B High cAMP C High cAMP & drug ( )


AC AC AC GPCR
GPCR GPCR

Gα12 Gα12 Gα12


PM PMGαs PMGαs
ATP cAMP ATP cAMP
C C
C
PDE
PDE PDE
RII RII

RII RII
RII RII

AKAP-lbc AKAP-lbc
P GEF AKAP-lbc
P GEF P GEF
14-3-3 14-3-3
PDE C 14-3-3 + PDE C + PDE C +

GDP- Rho Rho -GTP GDP- Rho Rho -GTP GDP- Rho Rho -GTP
PDE PDE PDE

Fig. (2). Targeting specific kinase substrates. Compartmentalized cross signaling between PKA and Rho signaling through the AKAP-lbc is
illustrated at conditions of low cAMP (A) where G12AKAP-RhoGEFRho signaling is undisturbed (rapid exchange of Rho(GDP) to
Rho(GTP)). (B) at high cAMP-levels (cAMP concentration gradient shown as gray shade, cAMP bound to the RII subunits shown as dark
gray), activated PKA can phosphorylate AKAP-Lbc (P enclosed by a circle), thereby providing a binding site for 14-3-3 proteins. Binding of
14-3-3 blocks access of Rho(GDP) to the RhoGEF domain, and thereby lowers Rho activation (formation of Rho(GTP)). Panel (C) shows
pharmacological intervention by a small molecule drug (black oval) that passively enters the cell (dotted reversible arrow) and binds the in-
terphase between 14-3-3 and PKA-phosphorylated AKAP-lbc. Binding of the drug further strengthen the interaction between 14-3-3 and
phospho-AKAP-lbs, thereby prolonging the PKA-mediated inhibition of Rho.
The cAMP-Dependent Protein Kinase Pathway as Therapeutic Target Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 1401

These are opportunities that are so far unexploited, but have specific drugs, simply because the same targets are used in
the potential of generating compounds that can discriminate different cells of the body. A possible solution to this can
at the substrate level. The screening method to detect prom- come from multi-targeting, e.g. by combining receptor or
ising compounds will however be more challenging than ligand manipulation with intracellular targets of the pathway
traditional kinase assays. of interest, to gain cell-type specificity. A synergistic interac-
tion between the different drugs would then enforce the
A specificity, making adverse side effects less pronounced. The
cAMP-signaling pathway provides many opportunities for
S such a strategy, extending from its coupling to multiple ex-
CATP CATP,S
tracellular receptor types, to the many downstream intracel-
Low affinity
lular targets such as kinases and their substrates. Perhaps
ATP ATP these opportunities of multi-targeting, and the range of dis-
S catalysis
C CS eases for which it bears relevance, can make the cAMP-
ADP Rate- ADP pathway a prototype for selective pharmacotargeting of cell
limiting signaling pathways.
High affinity ACKNOWLEDGEMENTS
CADP CADP,S
S This work was supported by the Norwegian Research
Council, the Norwegian Cancer Society and Western Nor-
pS way Regional Health Authority.

Inhibitor binding Mg/ATP ABBREVIATIONS


bound C-subunit
CATP,l1
B AC = Adenylyl cyclase

l1 tmAC = Transmembrane AC
S sAC = Soluble AC
CATP CATP,S
Low affinity AKAP = A-kinase anchoring protein
ATP ATP cNMP = Cyclic nucleotide monophoshate (either
S catalysis
C CS cAMP or cGMP)
ADP ADP CNS = Central nervous system
Exchange Exchange
stimulation, stimulation COPD = Chronic obstructive pulmonary disease
No substrate High affinity with substrate
CADP CADP,S CREB = cAMP response element binding protein
l2 S Epac = Exchange protein directly activated by cAMP
CADP,l2 pS GC = guanylyl cyclase
Inhibitor binding Mg/ADP Phosphorylated
bound C-subunit Substrate
ICER = Immediate cAMP-induced early repressor
PDE = Phosphodiesterase
PKA = cAMP dependent protein kinase
Fig. (3). PKA catalysis and opportunities for pharmacological
modulation. The kinetic mechanism for peptide phosphorylation is PKAI = PKA isotype I
shown (A) where the different ligand forms of C-subunit (CX) are PKAII = PKA isotype II
shown (X = ADP – Mg/ADP; = ATP – Mg/ATP; = ADP,S –
Mg/ADP+peptide; = ATP,S – Mg/ATP+peptide; = S – peptide). (B) PKG = cGMP-dependent protein kinase
shows targeting of different ligand-bound forms of C-subunit with ROCK = Rho-dependent protein kinase
inhibitors (I1) and (I2), binding to Mg/ATP and Mg/ADP-bound C-
subunit, respectively. Activators or inhibitors that modulate the rate REFERENCES
limiting nucleotide exchange are also shown. [1] Kim, C.; Cheng, C. Y.; Saldanha, S. A.; Taylor, S. S. PKA-I
holoenzyme structure reveals a mechanism for cAMP-dependent
CONCLUDING REMARKS activation. Cell, 2007, 130 (6), 1032-1043.
[2] Alverdi, V.; Mazon, H.; Versluis, C.; Hemrika, W.; Esposito, G.;
A major challenge for pharmacological manipulation of van den Heuvel, R.; Scholten, A.; Heck, A. J. cGMP-binding
prepares PKG for substrate binding by disclosing the C-terminal
physiological processes is a considerate level of similarity at domain. J. Mol. Biol., 2008, 375 (5), 1380-1393.
the molecular level, between different cell-types in the body. [3] Ellison, K. E.; Gandhi, G. Optimising the use of beta-adrenoceptor
This targeting dilemma is general, but perhaps at its most antagonists in coronary artery disease. Drugs, 2005, 65 (6), 787-
extreme in the CNS, where spatial confined signaling at the 797.
synapses mediates much of the specificity needed for the [4] McKeage, K.; Keam, S. J. Salmeterol/fluticasone propionate: a
review of its use in asthma. Drugs 2009, 69 (13), 1799-828.
communication of the neural networks. Side-effects can [5] Menendez, R.; Kelly, H. W. Theophylline therapy. J. Asthma,
therefore be expected during systemic exposure, even with 1983, 20 (6), 455-466.
1402 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 Kleppe et al.

[6] Boolell, M.; Allen, M. J.; Ballard, S. A.; Gepi-Attee, S.; Muirhead, expression and catalepsy in protein kinase A-deficient mice. Proc.
G. J.; Naylor, A. M.; Osterloh, I. H.; Gingell, C. Sildenafil: an Natl. Acad. Sci. USA, 1997, 94 (22), 12157-12161.
orally active type 5 cyclic GMP-specific phosphodiesterase [27] Uhler, M. D.; Chrivia, J. C.; McKnight, G. S. Evidence for a
inhibitor for the treatment of penile erectile dysfunction. Int. J. second isoform of the catalytic subunit of cAMP-dependent protein
Impot. Res., 1996, 8 (2), 47-52. kinase. J. Biol. Chem., 1986, 261 (33), 15360-15363.
[7] Jaski, B. E.; Fifer, M. A.; Wright, R. F.; Braunwald, E.; Colucci, [28] Showers, M. O.; Maurer, R. A. A cloned bovine cDNA encodes an
W. S. Positive inotropic and vasodilator actions of milrinone in alternate form of the catalytic subunit of cAMP-dependent protein
patients with severe congestive heart failure. Dose-response kinase. J. Biol. Chem., 1986, 261 (35), 16288-16291.
relationships and comparison to nitroprusside. J. Clin. Invest., [29] Beebe, S. J.; Oyen, O.; Sandberg, M.; Froysa, A.; Hansson, V.;
1985, 75 (2), 643-649. Jahnsen, T. Molecular cloning of a tissue-specific protein kinase (C
[8] Kamenetsky, M.; Middelhaufe, S.; Bank, E. M.; Levin, L. R.; gamma) from human testis--representing a third isoform for the
Buck, J.; Steegborn, C. Molecular details of cAMP generation in catalytic subunit of cAMP-dependent protein kinase. Mol.
mammalian cells: a tale of two systems. J. Mol. Biol., 2006, 362 Endocrinol., 1990, 4 (3), 465-475.
(4), 623-639. [30] Beebe, S. J.; Salomonsky, P.; Jahnsen, T.; Li, Y. The C gamma
[9] Cooper, D. M. Regulation and organization of adenylyl cyclases subunit is a unique isozyme of the cAMP-dependent protein kinase.
and cAMP. Biochem. J., 2003, 375 (Pt 3), 517-529. J. Biol. Chem., 1992, 267 (35), 25505-25512.
[10] Watson, P. A.; Giger, K. E.; Kempinski, A. M. Type I and type [31] Chiorini, J. A.; Zimmermann, B.; Yang, L.; Smith, R. H.; Ahearn,
VIII adenylyl cyclases constitute a family whose activation is A.; Herberg, F.; Kotin, R. M. Inhibition of PrKX, a novel protein
coupled to cellular deformation through the action of calcium- kinase, and the cyclic AMP-dependent protein kinase PKA by the
calmodulin. Biochem. Cell Biol., 1995, 73 (7-8), 367-372. regulatory proteins of adeno-associated virus type 2. Mol. Cell.
[11] Mittag, T. W.; Guo, W. B.; Kobayashi, K. Bicarbonate-activated Biol., 1998, 18 (10), 5921-5929.
adenylyl cyclase in fluid-transporting tissues. Am. J. Physiol., 1993, [32] Desseyn, J. L.; Burton, K. A.; McKnight, G. S. Expression of a
264 (6 Pt 2), F1060-1064. nonmyristylated variant of the catalytic subunit of protein kinase A
[12] Chen, Y.; Cann, M. J.; Litvin, T. N.; Iourgenko, V.; Sinclair, M. L.; during male germ-cell development. Proc. Natl. Acad. Sci. USA,
Levin, L. R.; Buck, J. Soluble adenylyl cyclase as an evolutionarily 2000, 97 (12), 6433-6438.
conserved bicarbonate sensor. Science, 2000, 289 (5479), 625-628. [33] Jedrzejewski, P. T.; Girod, A.; Tholey, A.; Konig, N.; Thullner, S.;
[13] Zaccolo, M.; Movsesian, M. A. cAMP and cGMP signaling cross- Kinzel, V.; Bossemeyer, D. A conserved deamidation site at Asn 2
talk: role of phosphodiesterases and implications for cardiac in the catalytic subunit of mammalian cAMP-dependent protein
pathophysiology. Circ. Res., 2007, 100 (11), 1569-1578. kinase detected by capillary LC-MS and tandem mass
[14] Movsesian, M.; Stehlik, J.; Vandeput, F.; Bristow, M. R. spectrometry. Protein Sci., 1998, 7 (2), 457-469.
Phosphodiesterase inhibition in heart failure. Heart Fail. Rev., [34] Kinzel, V.; Konig, N.; Pipkorn, R.; Bossemeyer, D.; Lehmann, W.
2009, 14 (4), 255-263. D. The amino terminus of PKA catalytic subunit--a site for
[15] Kleppisch, T. Phosphodiesterases in the central nervous system. introduction of posttranslational heterogeneities by deamidation: D-
Handb. Exp. Pharmacol., 2009, (191), 71-92. Asp2 and D-isoAsp2 containing isozymes. Protein Sci., 2000, 9
[16] Grant, P. G.; Colman, R. W. Purification and characterization of a (11), 2269-2277.
human platelet cyclic nucleotide phosphodiesterase. Biochemistry, [35] Skalhegg, B. S.; Huang, Y.; Su, T.; Idzerda, R. L.; McKnight, G.
1984, 23 (8), 1801-1807. S.; Burton, K. A. Mutation of the Calpha subunit of PKA leads to
[17] Heikaus, C. C.; Pandit, J.; Klevit, R. E. Cyclic nucleotide binding growth retardation and sperm dysfunction. Mol. Endocrinol., 2002,
GAF domains from phosphodiesterases: structural and mechanistic 16 (3), 630-639.
insights. Structure, 2009, 17 (12), 1551-1557. [36] Funderud, A.; Aas-Hanssen, K.; Aksaas, A. K.; Hafte, T. T.;
[18] Houslay, M. D.; Adams, D. R. PDE4 cAMP phosphodiesterases: Corthay, A.; Munthe, L. A.; Orstavik, S.; Skalhegg, B. S. Isoform-
modular enzymes that orchestrate signalling cross-talk, specific regulation of immune cell reactivity by the catalytic
desensitization and compartmentalization. Biochem. J., 2003, 370 subunit of protein kinase A (PKA). Cell Signal., 2009, 21 (2), 274-
(Pt 1), 1-18. 281.
[19] Takio, K.; Smith, S. B.; Krebs, E. G.; Walsh, K. A.; Titani, K. [37] Reinton, N.; Orstavik, S.; Haugen, T. B.; Jahnsen, T.; Tasken, K.;
Amino acid sequence of the regulatory subunit of bovine type II Skalhegg, B. S. A novel isoform of human cyclic 3',5'-adenosine
adenosine cyclic 3',5'-phosphate dependent protein kinase. monophosphate-dependent protein kinase, c alpha-s, localizes to
Biochemistry, 1984, 23 (18), 4200-4206. sperm midpiece. Biol. Reprod., 2000, 63 (2), 607-611.
[20] Titani, K.; Sasagawa, T.; Ericsson, L. H.; Kumar, S.; Smith, S. B.; [38] Orstavik, S.; Reinton, N.; Frengen, E.; Langeland, B. T.; Jahnsen,
Krebs, E. G.; Walsh, K. A. Amino acid sequence of the regulatory T.; Skalhegg, B. S. Identification of novel splice variants of the
subunit of bovine type I adenosine cyclic 3',5'-phosphate dependent human catalytic subunit Cbeta of cAMP-dependent protein kinase.
protein kinase. Biochemistry, 1984, 23 (18), 4193-4199. Eur. J. Biochem., 2001, 268 (19), 5066-5073.
[21] Ogreid, D.; Ekanger, R.; Suva, R. H.; Miller, J. P.; Sturm, P.; [39] Funderud, A.; Henanger, H. H.; Hafte, T. T.; Amieux, P. S.;
Corbin, J. D.; Doskeland, S. O. Activation of protein kinase Orstavik, S.; Skalhegg, B. S. Identification, cloning and
isozymes by cyclic nucleotide analogs used singly or in characterization of a novel 47 kDa murine PKA C subunit
combination. Principles for optimizing the isozyme specificity of homologous to human and bovine Cbeta2. BMC Biochem., 2006, 7,
analog combinations. Eur. J. Biochem., 1985, 150 (1), 219-227. 20.
[22] Francis, S. H.; Corbin, J. D. Cyclic nucleotide-dependent protein [40] Kvissel, A. K.; Orstavik, S.; Oistad, P.; Rootwelt, T.; Jahnsen, T.;
kinases: intracellular receptors for cAMP and cGMP action. Crit. Skalhegg, B. S. Induction of Cbeta splice variants and formation of
Rev. Clin. Lab. Sci., 1999, 36 (4), 275-328. novel forms of protein kinase A type II holoenzymes during
[23] Brandon, E. P.; Idzerda, R. L.; McKnight, G. S. PKA isoforms, retinoic acid-induced differentiation of human NT2 cells. Cell
neural pathways, and behaviour: making the connection. Curr. Signal., 2004, 16 (5), 577-587.
Opin. Neurobiol., 1997, 7 (3), 397-403. [41] Huang, Y.; Roelink, H.; McKnight, G. S. Protein kinase A
[24] Amieux, P. S.; Cummings, D. E.; Motamed, K.; Brandon, E. P.; deficiency causes axially localized neural tube defects in mice. J.
Wailes, L. A.; Le, K.; Idzerda, R. L.; McKnight, G. S. Biol. Chem., 2002, 277 (22), 19889-19896.
Compensatory regulation of RIalpha protein levels in protein [42] Howe, D. G.; Wiley, J. C.; McKnight, G. S. Molecular and
kinase A mutant mice. J. Biol. Chem., 1997, 272 (7), 3993-3998. behavioral effects of a null mutation in all PKA C beta isoforms.
[25] Brandon, E. P.; Zhuo, M.; Huang, Y. Y.; Qi, M.; Gerhold, K. A.; Mol. Cell. Neurosci., 2002, 20 (3), 515-524.
Burton, K. A.; Kandel, E. R.; McKnight, G. S.; Idzerda, R. L. [43] Zimmermann, B.; Chiorini, J. A.; Ma, Y.; Kotin, R. M.; Herberg, F.
Hippocampal long-term depression and depotentiation are defective W. PrKX is a novel catalytic subunit of the cAMP-dependent
in mice carrying a targeted disruption of the gene encoding the RI protein kinase regulated by the regulatory subunit type I. J. Biol.
beta subunit of cAMP-dependent protein kinase. Proc. Natl. Acad. Chem., 1999, 274 (9), 5370-5378.
Sci. USA, 1995, 92 (19), 8851-8855. [44] de Rooij, J.; Rehmann, H.; van Triest, M.; Cool, R. H.;
[26] Adams, M. R.; Brandon, E. P.; Chartoff, E. H.; Idzerda, R. L.; Wittinghofer, A.; Bos, J. L. Mechanism of regulation of the Epac
Dorsa, D. M.; McKnight, G. S. Loss of haloperidol induced gene family of cAMP-dependent RapGEFs. J. Biol. Chem., 2000, 275
(27), 20829-20836.
The cAMP-Dependent Protein Kinase Pathway as Therapeutic Target Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 1403

[45] de Rooij, J.; Zwartkruis, F. J.; Verheijen, M. H.; Cool, R. H.; adenosine 3',5'-cyclic monophosphothioate, the phosphothioate
Nijman, S. M.; Wittinghofer, A.; Bos, J. L. Epac is a Rap1 guanine- analogues of cAMP. Biochemistry, 2004, 43 (21), 6620-6629.
nucleotide-exchange factor directly activated by cyclic AMP. [65] Byeon, I. J.; Dao, K. K.; Jung, J.; Keen, J.; Leiros, I.; Doskeland, S.
Nature, 1998, 396 (6710), 474-477. O.; Martinez, A.; Gronenborn, A. M. Allosteric communication
[46] Niimura, M.; Miki, T.; Shibasaki, T.; Fujimoto, W.; Iwanaga, T.; between cAMP binding sites in the RI subunit of protein kinase A
Seino, S. Critical role of the N-terminal cyclic AMP-binding revealed by NMR. J. Biol. Chem., 2010, 285 (18), 14062-14070.
domain of Epac2 in its subcellular localization and function. J. [66] Kornev, A. P.; Taylor, S. S. Defining the conserved internal
Cell. Physiol., 2009, 219 (3), 652-658. architecture of a protein kinase. Biochim. Biophys. Acta, 2010,
[47] Ueno, H.; Shibasaki, T.; Iwanaga, T.; Takahashi, K.; Yokoyama, 1804 (3), 440-444.
Y.; Liu, L. M.; Yokoi, N.; Ozaki, N.; Matsukura, S.; Yano, H.; [67] Kopperud, R.; Krakstad, C.; Selheim, F.; Doskeland, S. O. cAMP
Seino, S. Characterization of the gene EPAC2: structure, effector mechanisms. Novel twists for an 'old' signaling system.
chromosomal localization, tissue expression, and identification of FEBS Lett., 2003, 546 (1), 121-126.
the liver-specific isoform. Genomics, 2001, 78 (1-2), 91-98. [68] Maeno, H.; Johnson, E. M.; Greengard, P. Subcellular distribution
[48] Rehmann, H.; Arias-Palomo, E.; Hadders, M. A.; Schwede, F.; of adenosine 3',5'-monophosphate-dependent protein kinase in rat
Llorca, O.; Bos, J. L. Structure of Epac2 in complex with a cyclic brain. J. Biol. Chem., 1971, 246 (1), 134-142.
AMP analogue and RAP1B. Nature, 2008, 455 (7209), 124-127. [69] Vallee, R. B.; DiBartolomeis, M. J.; Theurkauf, W. E. A protein
[49] Rehmann, H.; Das, J.; Knipscheer, P.; Wittinghofer, A.; Bos, J. L. kinase bound to the projection portion of MAP 2 (microtubule-
Structure of the cyclic-AMP-responsive exchange factor Epac2 in associated protein 2). J. Cell. Biol., 1981, 90 (3), 568-576.
its auto-inhibited state. Nature, 2006, 439 (7076), 625-628. [70] Carr, D. W.; Stofko-Hahn, R. E.; Fraser, I. D.; Bishop, S. M.;
[50] Ponsioen, B.; Gloerich, M.; Ritsma, L.; Rehmann, H.; Bos, J. L.; Acott, T. S.; Brennan, R. G.; Scott, J. D. Interaction of the
Jalink, K. Direct spatial control of Epac1 by cyclic AMP. Mol. regulatory subunit (RII) of cAMP-dependent protein kinase with
Cell. Biol., 2009, 29 (10), 2521-2531. RII-anchoring proteins occurs through an amphipathic helix
[51] Aumo, L.; Rusten, M.; Mellgren, G.; Bakke, M.; Lewis, A. E. binding motif. J. Biol. Chem., 1991, 266 (22), 14188-14192.
Functional roles of protein kinase A (PKA) and exchange protein [71] Kinderman, F. S.; Kim, C.; von Daake, S.; Ma, Y.; Pham, B. Q.;
directly activated by 3',5'-cyclic adenosine 5'-monophosphate Spraggon, G.; Xuong, N. H.; Jennings, P. A.; Taylor, S. S. A
(cAMP) 2 (EPAC2) in cAMP-mediated actions in adrenocortical dynamic mechanism for AKAP binding to RII isoforms of cAMP-
cells. Endocrinology, 2010, 151 (5), 2151-2161. dependent protein kinase. Mol. Cell., 2006, 24 (3), 397-408.
[52] Biel, M.; Michalakis, S. Cyclic nucleotide-gated channels. Handb. [72] Gold, M. G.; Lygren, B.; Dokurno, P.; Hoshi, N.; McConnachie,
Exp. Pharmacol., 2009, (191), 111-136. G.; Tasken, K.; Carlson, C. R.; Scott, J. D.; Barford, D. Molecular
[53] Fesenko, E. E.; Kolesnikov, S. S.; Lyubarsky, A. L. Induction by basis of AKAP specificity for PKA regulatory subunits. Mol. Cell.,
cyclic GMP of cationic conductance in plasma membrane of retinal 2006, 24 (3), 383-395.
rod outer segment. Nature, 1985, 313 (6000), 310-313. [73] Newlon, M. G.; Roy, M.; Morikis, D.; Hausken, Z. E.; Coghlan,
[54] Distler, M.; Biel, M.; Flockerzi, V.; Hofmann, F. Expression of V.; Scott, J. D.; Jennings, P. A. The molecular basis for protein
cyclic nucleotide-gated cation channels in non-sensory tissues and kinase A anchoring revealed by solution NMR. Nat. Struct. Biol.,
cells. Neuropharmacology, 1994, 33 (11), 1275-1282. 1999, 6 (3), 222-227.
[55] Kingston, P. A.; Zufall, F.; Barnstable, C. J. Rat hippocampal [74] Pidoux, G.; Tasken, K. Specificity and spatial dynamics of protein
neurons express genes for both rod retinal and olfactory cyclic kinase A signaling organized by A-kinase-anchoring proteins. J.
nucleotide-gated channels: novel targets for cAMP/cGMP function. Mol. Endocrinol., 2010, 44 (5), 271-284.
Proc. Natl. Acad. Sci. USA, 1996, 93 (19), 10440-10445. [75] Lim, C. J.; Han, J.; Yousefi, N.; Ma, Y.; Amieux, P. S.; McKnight,
[56] Cadd, G. G.; Uhler, M. D.; McKnight, G. S. Holoenzymes of G. S.; Taylor, S. S.; Ginsberg, M. H. Alpha4 integrins are type I
cAMP-dependent protein kinase containing the neural form of type cAMP-dependent protein kinase-anchoring proteins. Nat. Cell
I regulatory subunit have an increased sensitivity to cyclic Biol., 2007, 9 (4), 415-421.
nucleotides. J. Biol. Chem., 1990, 265 (32), 19502-19506. [76] Huang, L. J.; Durick, K.; Weiner, J. A.; Chun, J.; Taylor, S. S.
[57] Dostmann, W. R.; Taylor, S. S. Identifying the molecular switches Identification of a novel protein kinase A anchoring protein that
that determine whether (Rp)-cAMPS functions as an antagonist or binds both type I and type II regulatory subunits. J. Biol. Chem.,
an agonist in the activation of cAMP-dependent protein kinase I. 1997, 272 (12), 8057-8064.
Biochemistry, 1991, 30 (35), 8710-8716. [77] Kapiloff, M. S.; Piggott, L. A.; Sadana, R.; Li, J.; Heredia, L. A.;
[58] Kopperud, R.; Christensen, A. E.; Kjarland, E.; Viste, K.; Kleivdal, Henson, E.; Efendiev, R.; Dessauer, C. W. An adenylyl cyclase-
H.; Doskeland, S. O. Formation of inactive cAMP-saturated mAKAPbeta signaling complex regulates cAMP levels in cardiac
holoenzyme of cAMP-dependent protein kinase under myocytes. J. Biol. Chem., 2009, 284 (35), 23540-23546.
physiological conditions. J. Biol. Chem., 2002, 277 (16), 13443- [78] Di Benedetto, G.; Zoccarato, A.; Lissandron, V.; Terrin, A.; Li, X.;
13448. Houslay, M. D.; Baillie, G. S.; Zaccolo, M. Protein kinase A type I
[59] Viste, K.; Kopperud, R. K.; Christensen, A. E.; Doskeland, S. O. and type II define distinct intracellular signaling compartments.
Substrate enhances the sensitivity of type I protein kinase a to Circ. Res., 2008, 103 (8), 836-844.
cAMP. J. Biol. Chem., 2005, 280 (14), 13279-13284. [79] Zaccolo, M.; Di Benedetto, G.; Lissandron, V.; Mancuso, L.;
[60] Dao, K. K.; Teigen, K.; Kopperud, R.; Hodneland, E.; Schwede, F.; Terrin, A.; Zamparo, I. Restricted diffusion of a freely diffusible
Christensen, A. E.; Martinez, A.; Doskeland, S. O. Epac1 and second messenger: mechanisms underlying compartmentalized
cAMP-dependent protein kinase holoenzyme have similar cAMP cAMP signalling. Biochem. Soc. Trans., 2006, 34 (Pt 4), 495-497.
affinity, but their cAMP domains have distinct structural features [80] DeLange, R. J.; Kemp, R. G.; Riley, W. D.; Cooper, R. A.; Krebs,
and cyclic nucleotide recognition. J. Biol. Chem., 2006, 281 (30), E. G. Activation of skeletal muscle phosphorylase kinase by
21500-21511. adenosine triphosphate and adenosine 3',5'-monophosphate. J. Biol.
[61] Wu, J.; Brown, S. H.; von Daake, S.; Taylor, S. S. PKA type Chem., 1968, 243 (9), 2200-2208.
IIalpha holoenzyme reveals a combinatorial strategy for isoform [81] Petersen, R. K.; Madsen, L.; Pedersen, L. M.; Hallenborg, P.;
diversity. Science, 2007, 318 (5848), 274-279. Hagland, H.; Viste, K.; Doskeland, S. O.; Kristiansen, K. Cyclic
[62] McNicholl, E. T.; Das, R.; Sildas, S.; Taylor, S. S.; Melacini, G. AMP (cAMP)-mediated stimulation of adipocyte differentiation
Communication between tandem cAMP-binding domains in the requires the synergistic action of Epac- and cAMP-dependent
regulatory subunit of protein kinase A (PKA)-I{alpha} as revealed protein kinase-dependent processes. Mol. Cell Biol., 2008, 28 (11),
by domain-silencing mutations. J. Biol. Chem., 2010, 285 (20), 3804-3816.
15523-15537. [82] Zhao, A. Z. Control of food intake through regulation of cAMP.
[63] Kim, C.; Xuong, N. H.; Taylor, S. S. Crystal structure of a complex Curr. Top. Dev. Biol., 2005, 67, 207-224.
between the catalytic and regulatory (RIalpha) subunits of PKA. [83] Wang, Z.; Li, V.; Chan, G. C.; Phan, T.; Nudelman, A. S.; Xia, Z.;
Science, 2005, 307 (5710), 690-696. Storm, D. R. Adult type 3 adenylyl cyclase-deficient mice are
[64] Wu, J.; Jones, J. M.; Nguyen-Huu, X.; Ten Eyck, L. F.; Taylor, S. obese. PLoS One, 2009, 4 (9), e6979.
S. Crystal structures of RIalpha subunit of cyclic adenosine 5'- [84] Cummings, D. E.; Brandon, E. P.; Planas, J. V.; Motamed, K.;
monophosphate (cAMP)-dependent protein kinase complexed with Idzerda, R. L.; McKnight, G. S. Genetically lean mice result from
(Rp)-adenosine 3',5'-cyclic monophosphothioate and (Sp)-
1404 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 Kleppe et al.

targeted disruption of the RII beta subunit of protein kinase A. [105] Almas, B.; Le Bourdelles, B.; Flatmark, T.; Mallet, J.; Haavik, J.
Nature, 1996, 382 (6592), 622-626. Regulation of recombinant human tyrosine hydroxylase isozymes
[85] Newhall, K. J.; Cummings, D. E.; Nolan, M. A.; McKnight, G. S. by catecholamine binding and phosphorylation. Structure/activity
Deletion of the RIIbeta-subunit of protein kinase A decreases body studies and mechanistic implications. Eur. J. Biochem., 1992, 209
weight and increases energy expenditure in the obese, leptin- (1), 249-255.
deficient ob/ob mouse. Mol. Endocrinol., 2005, 19 (4), 982-991. [106] Winge, I.; McKinney, J. A.; Ying, M.; D'Santos, C. S.; Kleppe, R.;
[86] Metrich, M.; Lucas, A.; Gastineau, M.; Samuel, J. L.; Heymes, C.; Knappskog, P. M.; Haavik, J. Activation and stabilization of human
Morel, E.; Lezoualc'h, F. Epac mediates beta-adrenergic receptor- tryptophan hydroxylase 2 by phosphorylation and 14-3-3 binding.
induced cardiomyocyte hypertrophy. Circ. Res., 2008, 102 (8), Biochem. J., 2008, 410 (1), 195-204.
959-965. [107] Zhang, H. T. Cyclic AMP-specific phosphodiesterase-4 as a target
[87] Bogatcheva, N. V.; Zemskova, M. A.; Kovalenkov, Y.; Poirier, C.; for the development of antidepressant drugs. Curr. Pharm. Des.,
Verin, A. D. Molecular mechanisms mediating protective effect of 2009, 15 (14), 1688-1698.
cAMP on lipopolysaccharide (LPS)-induced human lung [108] Bourgault, S.; Vaudry, D.; Dejda, A.; Doan, N. D.; Vaudry, H.;
microvascular endothelial cells (HLMVEC) hyperpermeability. J. Fournier, A. Pituitary adenylate cyclase-activating polypeptide:
Cell Physiol., 2009, 221 (3), 750-759. focus on structure-activity relationships of a neuroprotective
[88] Aslam, M.; Hartel, F. V.; Arshad, M.; Gunduz, D.; Abdallah, Y.; Peptide. Curr. Med. Chem., 2009, 16 (33), 4462-4480.
Sauer, H.; Piper, H. M.; Noll, T. cAMP/PKA antagonizes [109] Shelton, R. C.; Hal Manier, D.; Lewis, D. A. Protein kinases A and
thrombin-induced inactivation of endothelial myosin light chain C in post-mortem prefrontal cortex from persons with major
phosphatase: role of CPI-17. Cardiovasc. Res., 2010, 87 (2), 375- depression and normal controls. Int. J. Neuropsychopharmacol.,
384. 2009, 12 (9), 1223-1232.
[89] Birukova, A. A.; Birukov, K. G.; Adyshev, D.; Usatyuk, P.; [110] Dwivedi, Y.; Rizavi, H. S.; Shukla, P. K.; Lyons, J.; Faludi, G.;
Natarajan, V.; Garcia, J. G.; Verin, A. D. Involvement of Palkovits, M.; Sarosi, A.; Conley, R. R.; Roberts, R. C.;
microtubules and Rho pathway in TGF-beta1-induced lung Tamminga, C. A.; Pandey, G. N. Protein kinase A in postmortem
vascular barrier dysfunction. J. Cell Physiol., 2005, 204 (3), 934- brain of depressed suicide victims: altered expression of specific
947. regulatory and catalytic subunits. Biol. Psychiatry, 2004, 55 (3),
[90] Sehrawat, S.; Cullere, X.; Patel, S.; Italiano, J. Jr.; Mayadas, T. N. 234-243.
Role of Epac1, an exchange factor for Rap GTPases, in endothelial [111] Liang, Z.; Liu, F.; Grundke-Iqbal, I.; Iqbal, K.; Gong, C. X. Down-
microtubule dynamics and barrier function. Mol. Biol. Cell, 2008, regulation of cAMP-dependent protein kinase by over-activated
19 (3), 1261-1270. calpain in Alzheimer disease brain. J. Neurochem., 2007, 103 (6),
[91] Surapisitchat, J.; Jeon, K. I.; Yan, C.; Beavo, J. A. Differential 2462-2470.
regulation of endothelial cell permeability by cGMP via [112] Peters-Golden, M. Putting on the brakes: cyclic AMP as a
phosphodiesterases 2 and 3. Circ. Res., 2007, 101 (8), 811-818. multipronged controller of macrophage function. Sci. Signal., 2009,
[92] Birukova, A. A.; Zagranichnaya, T.; Alekseeva, E.; Bokoch, G. M.; 2 (75), pe37.
Birukov, K. G. Epac/Rap and PKA are novel mechanisms of ANP- [113] Wall, E. A.; Zavzavadjian, J. R.; Chang, M. S.; Randhawa, B.; Zhu,
induced Rac-mediated pulmonary endothelial barrier protection. J. X.; Hsueh, R. C.; Liu, J.; Driver, A.; Bao, X. R.; Sternweis, P. C.;
Cell Physiol., 2008, 215 (3), 715-724. Simon, M. I.; Fraser, I. D. Suppression of LPS-induced TNF-alpha
[93] Hatzelmann, A.; Morcillo, E. J.; Lungarella, G.; Adnot, S.; Sanjar, production in macrophages by cAMP is mediated by PKA-
S.; Beume, R.; Schudt, C.; Tenor, H. The preclinical pharmacology AKAP95-p105. Sci. Signal, 2009, 2 (75), ra28.
of roflumilast - A selective, oral phosphodiesterase 4 inhibitor in [114] Bryn, T.; Mahic, M.; Enserink, J. M.; Schwede, F.; Aandahl, E. M.;
development for chronic obstructive pulmonary disease. Pulm Tasken, K. The cyclic AMP-Epac1-Rap1 pathway is dissociated
Pharmacol. Ther., 2010, 23 (4), 235-256. from regulation of effector functions in monocytes but acquires
[94] Schwarz, U. R.; Walter, U.; Eigenthaler, M. Taming platelets with immunoregulatory function in mature macrophages. J. Immunol.,
cyclic nucleotides. Biochem. Pharmacol., 2001, 62 (9), 1153-1161. 2006, 176 (12), 7361-7370.
[95] Fabre, J. E.; Gurney, M. E. Limitations of current therapies to [115] Agarwal, N.; Lamichhane, G.; Gupta, R.; Nolan, S.; Bishai, W. R.
prevent thrombosis: a need for novel strategies. Mol. Biosyst., Cyclic AMP intoxication of macrophages by a Mycobacterium
2010, 6 (2), 305-315. tuberculosis adenylate cyclase. Nature, 2009, 460 (7251), 98-102.
[96] Colman, R. W. Platelet cyclic adenosine monophosphate [116] Torgersen, K. M.; Aandahl, E. M.; Tasken, K. Molecular
phosphodiesterases: targets for regulating platelet-related architecture of signal complexes regulating immune cell function.
thrombosis. Semin. Thromb. Hemost., 2004, 30 (4), 451-460. Handb. Exp. Pharmacol., 2008, (186), 327-363.
[97] Haslam, R. J.; Dickinson, N. T.; Jang, E. K. Cyclic nucleotides and [117] Mattapallil, J. J.; Douek, D. C.; Hill, B.; Nishimura, Y.; Martin, M.;
phosphodiesterases in platelets. Thromb. Haemost., 1999, 82 (2), Roederer, M. Massive infection and loss of memory CD4+ T cells
412-423. in multiple tissues during acute SIV infection. Nature, 2005, 434
[98] Jensen, B. O.; Selheim, F.; Doskeland, S. O.; Gear, A. R.; (7037), 1093-1097.
Holmsen, H. Protein kinase A mediates inhibition of the thrombin- [118] Barnitz, R. A.; Wan, F.; Tripuraneni, V.; Bolton, D. L.; Lenardo,
induced platelet shape change by nitric oxide. Blood, 2004, 104 (9), M. J. Protein Kinase A phosphorylation activates Vpr-induced cell
2775-2782. cycle arrest during Human Immunodeficiency Virus Type-1
[99] Schafe, G. E.; LeDoux, J. E. Memory consolidation of auditory infection. J. Virol., 2010, 84 (13), 6410-6424.
pavlovian fear conditioning requires protein synthesis and protein [119] Nokta, M. A.; Pollard, R. B. Human immunodeficiency virus
kinase A in the amygdala. J. Neurosci., 2000, 20 (18), RC96. replication: modulation by cellular levels of cAMP. AIDS Res.
[100] Kaplan, M. P.; Abel, T. Genetic approaches to the study of synaptic Hum. Retroviru., 1992, 8 (7), 1255-1261.
plasticity and memory storage. CNS Spectr., 2003, 8 (8), 597-610. [120] Mongillo, M.; Tocchetti, C. G.; Terrin, A.; Lissandron, V.; Cheung,
[101] Malleret, G.; Alarcon, J. M.; Martel, G.; Takizawa, S.; Vronskaya, Y. F.; Dostmann, W. R.; Pozzan, T.; Kass, D. A.; Paolocci, N.;
S.; Yin, D.; Chen, I. Z.; Kandel, E. R.; Shumyatsky, G. P. Houslay, M. D.; Zaccolo, M. Compartmentalized
Bidirectional regulation of hippocampal long-term synaptic phosphodiesterase-2 activity blunts beta-adrenergic cardiac
plasticity and its influence on opposing forms of memory. J. inotropy via an NO/cGMP-dependent pathway. Circ. Res., 2006,
Neurosci., 2010, 30 (10), 3813-25. 98 (2), 226-234.
[102] Lee, H. K.; Barbarosie, M.; Kameyama, K.; Bear, M. F.; Huganir, [121] Lin, D. T.; Fretier, P.; Jiang, C.; Vincent, S. R. Nitric oxide
R. L. Regulation of distinct AMPA receptor phosphorylation sites signaling via cGMP-stimulated phosphodiesterase in striatal
during bidirectional synaptic plasticity. Nature, 2000, 405 (6789), neurons. Synapse, 2010, 64 (6), 460-466.
955-959. [122] Jager, R.; Schwede, F.; Genieser, H. G.; Koesling, D.; Russwurm,
[103] Sealfon, S. C.; Olanow, C. W. Dopamine receptors: from structure M. Activation of PDE2 and PDE5 by specific GAF ligands:
to behavior. Trends Neurosci., 2000, 23 (10 Suppl), S34-40. delayed activation of PDE5. Br J Pharmacol., 2010, 161 (7), 1645-
[104] Millan, M. J.; Marin, P.; Bockaert, J.; la Cour, C. M. Signaling at 1660.
G-protein-coupled serotonin receptors: recent advances and future [123] Pierre, S.; Eschenhagen, T.; Geisslinger, G.; Scholich, K. Capturing
research directions. Trends Pharmacol. Sci., 2008, 29 (9), 454-464. adenylyl cyclases as potential drug targets. Nat. Rev. Drug Discov.,
2009, 8 (4), 321-335.
The cAMP-Dependent Protein Kinase Pathway as Therapeutic Target Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 11 1405

[124] Kikura, M.; Morita, K.; Sato, S. Pharmacokinetics and a simulation [135] Bogoyevitch, M. A.; Fairlie, D. P. A new paradigm for protein
model of colforsin daropate, new forskolin derivative inotropic kinase inhibition: blocking phosphorylation without directly
vasodilator, in patients undergoing coronary artery bypass grafting. targeting ATP binding. Drug Discov. Today, 2007, 12 (15-16),
Pharmacol. Res., 2004, 49 (3), 275-281. 622-633.
[125] Buck, J.; Levin, L. R.; Mühlschlegel, F. A. Chemical inhibitors of [136] Shomin, C. D.; Meyer, S. C.; Ghosh, I. Staurosporine tethered
soluble Adeneylyl cyclase (SAC). Patent Number 2005, peptide ligands that target cAMP-dependent protein kinase (PKA):
WO/2005/070419. optimization and selectivity profiling. Bioorg. Med. Chem., 2009,
[126] Christensen, A. E.; Selheim, F.; de Rooij, J.; Dremier, S.; Schwede, 17 (17), 6196-6202.
F.; Dao, K. K.; Martinez, A.; Maenhaut, C.; Bos, J. L.; Genieser, H. [137] Carlson, C. R.; Lygren, B.; Berge, T.; Hoshi, N.; Wong, W.;
G.; Doskeland, S. O. cAMP analog mapping of Epac1 and cAMP Tasken, K.; Scott, J. D. Delineation of type I protein kinase A-
kinase. Discriminating analogs demonstrate that Epac and cAMP selective signaling events using an RI anchoring disruptor. J. Biol.
kinase act synergistically to promote PC-12 cell neurite extension. Chem., 2006, 281 (30), 21535-21545.
J. Biol. Chem., 2003, 278 (37), 35394-35402. [138] Torheim, E. A.; Jarnaess, E.; Lygren, B.; Tasken, K. Design of
[127] Ogreid, D.; Ekanger, R.; Suva, R. H.; Miller, J. P.; Doskeland, S. proteolytically stable RI-anchoring disruptor peptidomimetics for
O. Comparison of the two classes of binding sites (A and B) of type in vivo studies of anchored type I protein kinase A-mediated
I and type II cyclic-AMP-dependent protein kinases by using cyclic signalling. Biochem. J., 2009, 424 (1), 69-78.
nucleotide analogs. Eur. J. Biochem., 1989, 181 (1), 19-31. [139] Morrison, D. K. The 14-3-3 proteins: integrators of diverse
[128] Rehmann, H.; Schwede, F.; Doskeland, S. O.; Wittinghofer, A.; signaling cues that impact cell fate and cancer development. Trends
Bos, J. L. Ligand-mediated activation of the cAMP-responsive Cell Biol., 2009, 19 (1), 16-23.
guanine nucleotide exchange factor Epac. J. Biol. Chem., 2003, 278 [140] Meiri, D.; Greeve, M. A.; Brunet, A.; Finan, D.; Wells, C. D.;
(40), 38548-38556. LaRose, J.; Rottapel, R. Modulation of Rho guanine exchange
[129] Birkeland, E.; Nygaard, G.; Oveland, E.; Mjaavatten, O.; Ljones, factor Lfc activity by protein kinase A-mediated phosphorylation.
M.; Doskeland, S. O.; Krakstad, C.; Selheim, F. Epac-induced Mol. Cell. Biol., 2009, 29 (21), 5963-5973.
Alterations in the Proteome of Human SH-SY5Y Neuroblastoma [141] Diviani, D.; Abuin, L.; Cotecchia, S.; Pansier, L. Anchoring of both
Cells. J. Proteomics Bioinform., 2009, 2 (6), 244-254. PKA and 14-3-3 inhibits the Rho-GEF activity of the AKAP-Lbc
[130] Laxman, S.; Riechers, A.; Sadilek, M.; Schwede, F.; Beavo, J. A. signaling complex. EMBO J., 2004, 23 (14), 2811-2820.
Hydrolysis products of cAMP analogs cause transformation of [142] Wurtele, M.; Jelich-Ottmann, C.; Wittinghofer, A.; Oecking, C.
Trypanosoma brucei from slender to stumpy-like forms. Proc. Natl. Structural view of a fungal toxin acting on a 14-3-3 regulatory
Acad. Sci. USA, 2006, 103 (50), 19194-19199. complex. EMBO J., 2003, 22 (5), 987-994.
[131] Poppe, H.; Rybalkin, S. D.; Rehmann, H.; Hinds, T. R.; Tang, X. [143] Whitehouse, S.; Feramisco, J. R.; Casnellie, J. E.; Krebs, E. G.;
B.; Christensen, A. E.; Schwede, F.; Genieser, H. G.; Bos, J. L.; Walsh, D. A. Studies on the kinetic mechanism of the catalytic
Doskeland, S. O.; Beavo, J. A.; Butt, E. Cyclic nucleotide analogs subunit of the cAMP-dependent protein kinase. J. Biol. Chem.,
as probes of signaling pathways. Nat. Methods, 2008, 5 (4), 277- 1983, 258 (6), 3693-3701.
278. [144] Petukhov, S. P.; Grivennikov, I. A.; Bulargina, T. V.; Severin, E. S.
[132] Liu, Y.; Gray, N. S. Rational design of inhibitors that bind to [Kinetic mechanism of phosphotransferase reactions catalyzed by
inactive kinase conformations. Nat. Chem. Biol., 2006, 2 (7), 358- cAMP-dependent protein kinase type I and type II from rabbit
364. skeletal muscle]. Biokhimiia, 1984, 49 (8), 1367-1374.
[133] Wong, S.; Witte, O. N. The BCR-ABL story: bench to bedside and [145] Zhou, J.; Adams, J. A. Participation of ADP dissociation in the
back. Annu. Rev. Immunol., 2004, 22, 247-306. rate-determining step in cAMP-dependent protein kinase.
[134] Davies, S. P.; Reddy, H.; Caivano, M.; Cohen, P. Specificity and Biochemistry, 1997, 36 (50), 15733-15738.
mechanism of action of some commonly used protein kinase [146] Shaffer, J.; Adams, J. A. Detection of conformational changes
inhibitors. Biochem. J., 2000, 351 (Pt 1), 95-105. along the kinetic pathway of protein kinase A using a catalytic
trapping technique. Biochemistry, 1999, 38 (37), 12072-12079.

Received: June 01, 2010 Revised: August 13, 2010 Accepted: September 13, 2010

You might also like