Tải bản đầy đủ (.pdf) (8 trang)

Tài liệu Báo cáo khoa học: Second messenger function and the structure–activity relationship of cyclic adenosine diphosphoribose (cADPR) doc

Bạn đang xem bản rút gọn của tài liệu. Xem và tải ngay bản đầy đủ của tài liệu tại đây (254.86 KB, 8 trang )

MINIREVIEW
Second messenger function and the structure–activity
relationship of cyclic adenosine diphosphoribose (cADPR)
Andreas H. Guse
University Medical Center Hamburg-Eppendorf, Center of Experimental Medicine, Institute of Biochemistry and Molecular Biology I,
Cellular Signal Transduction, Hamburg, Germany
The cyclic ADP-ribose ⁄ Ca
2+
signalling
pathway
Cyclic ADP-ribose (cADPR) was discovered in 1987 as
aCa
2+
mobilizing metabolite of the well-known co-
enzyme b-nicotinamide adenine dinucleotide (NAD) by
Lee and coworkers [1]. The cyclic structure of cADPR
was initially predicted to originate from an N-glycosyl
linkage between the anomeric carbon of the ribose,
which in the precursor NAD is linked to nicotinamide,
and the amino ⁄ imino group at C6 of the adenine
moiety [2]. Spectroscopic data [3] and finally a crystal
structure revealed cyclization between the anomeric
C1 of this ribose moiety (commonly termed ‘northern
ribose’ while the ribose linked to N9 of adenine is
called the ‘southern’ ribose; Fig. 1) and the N1 of the
adenine ring [4].
Besides d-myo-inositol 1,4,5-trisphosphate (InsP
3
)
and nicotinic acid adenine dinucleotide phosphate
(NAADP; reviewed in [4a]), cADPR is one of the prin-


cipal Ca
2+
-releasing second messengers involved in cel-
lular Ca
2+
homeostasis. Changes in the cellular Ca
2+
homeostasis are among the fundamental signalling pro-
cesses in multicellular organisms. Such changes occur
in response to extracellular signals, e.g. hormones,
mediators, cell–cell contacts or physical stimuli, and
represent one of the most important, powerful and ver-
satile intracellular signal transducers. Changes in the
Correspondence
A. H. Guse, University Medical Center
Hamburg-Eppendorf, Center of Experimental
Medicine, Institute of Biochemistry and
Molecular Biology I: Cellular Signal
Transduction, Martinistr. 52,
20246 Hamburg, Germany
Fax: +49 40 42803 9880
Tel: +49 40 42803 2828
E-mail:
(Received 10 March 2005, accepted 05 July
2005)
doi:10.1111/j.1742-4658.2005.04863.x
Cyclic ADP-ribose (cADPR) is a Ca
2+
mobilizing second messenger found
in various cell types, tissues and organisms. Receptor-mediated formation

of cADPR may proceed via transmembrane shuttling of the substrate
NAD and involvement of the ectoenzyme CD38, or via so far unidentified
ADP-ribosyl cyclases located within the cytosol or in internal membranes.
cADPR activates intracellular Ca
2+
release via type 2 and 3 ryanodine
receptors. The exact molecular mechanism, however, remains to be elucida-
ted. Possibilities are the direct binding of cADPR to the ryanodine receptor
or binding via a separate cADPR binding protein. In addition to Ca
2+
release, cADPR also evokes Ca
2+
entry. The underlying mechanism(s) may
comprise activation of capacitative Ca
2+
entry and ⁄ or activation of the
cation channel TRPM2 in conjunction with adenosine diphosphoribose.
The development of novel cADPR analogues revealed new insights into the
structure–activity relationship. Substitution of either the northern ribose or
both the northern and southern ribose resulted in much simpler molecules,
which still retained significant biological activity.
Abbreviations
ADPRC, ADP-ribosyl cyclase; 8-Br-N1-cIDPR, 8-bromo-cyclic inosine diphosphoribose; cADPcR, cyclic ADP carbocyclic ribose; cADPR, cyclic
adenosine diphosphoribose; cADPR-BP, cADPR binding protein; cArisDPR, cyclic aristeromycin diphosphoribose; N1-cIDPR, N1-coupled
cyclic inosine diphosphoribose; cIDP-DE, N1-[(phosphoryl-O-ethoxy)-methyl]-N9-[(phosphoryl-O-ethoxy)-methyl]-hypoxanthine-cyclic pyro-
phosphate; cIDPRE, N1-ethoxymethyl-cIDPR; CRAC, Ca
2+
release activated Ca
2+
channel; FKBP, FK506 binding protein; InsP

3
, D-myo-inositol
1,4,5-trisphosphate; NAADP, nicotinic acid adenine dinucleotide phosphate; RyR, ryanodine receptor; TRP, transient receptor potential.
4590 FEBS Journal 272 (2005) 4590–4597 ª 2005 FEBS
cellular Ca
2+
homeostasis finally result in meaningful
physiological response of the cell. Thus, intracellular
Ca
2+
signalling is one of the most important transduc-
tion systems to integrate physiological responses of
multicellular organisms.
Because the free cytosolic and nuclear Ca
2+
concen-
tration ([Ca
2+
]
i
) is kept fairly low (approximately
50–100 nm) by ATP-driven Ca
2+
pumps located in
both the plasma membrane and intracellular mem-
branes (reviewed in [5]), rapid increases of [Ca
2+
]
i
can

be achieved by increasing the open probability of
Ca
2+
channels, either localized in the membranes of
intracellular Ca
2+
stores or in the plasma membrane.
Such Ca
2+
entry channels in the plasma membrane
and Ca
2+
release channels in intracellular membranes
have been reviewed in the past [6–11]. Review articles
dealing with the cADPR ⁄ Ca
2+
signalling system, the
topic of this article, have also been published in the
last two years [12–16]. Thus, I will not repeat in detail
the topics presented in those reviews, but I will briefly
describe the hallmarks of the cADPR ⁄ Ca
2+
signalling
system. Subsequently I will spend more time in discuss-
ing recent findings related to the biological activity of
cADPR analogues and some clues regarding the struc-
ture–activity relationship of cADPR.
The cADPR ⁄ Ca
2+
signalling system is active in

diverse cellular systems, including smooth, skeletal and
cardiac muscle, neuronal and neuronal-related cells,
hemopoietic cells, acinar cells, and oocytes (for a more
complete list see [15]). Because the cADPR ⁄ Ca
2+
sig-
nalling system was also observed in protozoa and plant
cells, it appears to be a phylogenetically old and con-
served system. As for the InsP
3
⁄ Ca
2+
signalling sys-
tem, in several cell types extracellular stimuli activate
cADPR-forming enzymes called ADP-ribosyl cyclases
Fig. 2. Receptor-mediated formation, metabolism and sites of action of cADPR. Dotted lines indicate minor pathway or relations not gener-
ally accepted or proven. Cx43, connexin 43; CRAC, Ca
2+
release-activated Ca
2+
channel; cADPRH, cADPR-hydrolase.
Fig. 1. Structure of cADPR.
A. H. Guse Second messenger function of cADPR
FEBS Journal 272 (2005) 4590–4597 ª 2005 FEBS 4591
(ADPRC) and thereby induce the formation of
cADPR (Fig. 2). G-protein coupling and Tyr phos-
phorylation have been implicated in ADPRC activa-
tion [17,18].
The enzymes responsible for the synthesis of cADPR
are still a matter of debate. An ADPRC that acts

mainly as a cyclizing enzyme has been purified and
cloned more than 10 years ago from the ovotestis of
Aplysia californica [19,20]. Mammalian homologues of
this enzyme are the membrane proteins CD38 and
CD157 (reviewed in [21]). After their discovery it was
surprising to note that their catalytic sites are located
outside of the cell (or in intracellular vesicles), but
obviously not in direct contact to the substrate NAD
and the intracellular Ca
2+
release channel sensitive to
cADPR, the ryanodine receptor (RyR). This situation
has been described as the ‘topological paradox’ of the
cADPR ⁄ Ca
2+
signalling system [22]. De Flora and
coworkers have worked out a potential solution for
this problem. They found that NAD can leave the cell
via connexin 43 hemichannels (Fig. 2; [23]). Outside
the cell (or inside CD38 containing vesicles) NAD is
then converted, at least in part, to cADPR. Evidence
was presented that both CD38 and nucleoside trans-
porters act as cADPR-transporting proteins (Fig. 2;
[24]). This system in principle represents a solution for
the topological paradox. However, connexin 43 hemi-
channels appear to be open for NAD export only at
[Ca
2+
]
i

 100 nm, indicating that this system is un-
likely to operate when [Ca
2+
]
i
is elevated above nor-
mal basal levels [25]. On the other hand, ectoenzymes
producing cADPR (such as CD38 and CD157) and
transport systems for cADPR in the plasma membrane
open the possibility that cADPR acts as a paracrine
signalling molecule (reviewed in [14]). Indeed, recently
a potentially very important example for such a para-
crine intercellular signalling molecule was described.
CD157-(BST-1)-positive bone marrow stromal cells via
production of extracellular cADPR induced the expan-
sion of human hemopoietic progenitor cells [26]. A
crucial point in this intercellular signalling pathway
appears to be the expression of concentrative nucleo-
side transporters in the hemopoietic progenitors since
this allows uptake of a sufficient amount of cADPR
into the target cells [26].
In addition to the relatively complicated system for
cADPR synthesis described above, several reports sug-
gest expression of either cytosolic or membrane-bound
enzymes not related to CD38 or CD157 [18,27–31].
None of these enzymes have been identified on the
molecular level so far; however, some (or all) of them
might be located at cellular sites more suitable for
rapid formation of intracellular cADPR.
Ca

2+
release by cADPR via ryanodine
receptors
Whatever these enzymes turn out to be, receptor-medi-
ated formation of cADPR obviously takes place in
many cell types and cADPR acts on the type 2 and ⁄ or
type 3 RyR. This interaction was initially demonstra-
ted by the sensitivity of cADPR-mediated Ca
2+
release
to pharmacological inhibitors of RyR, such as ruthen-
ium red or inhibitory concentrations of ryanodine [32]
and has since been confirmed in many cell systems.
Moreover, molecular knock-down of type 3 RyR in
T-lymphocytes resulted in a significant reduction of
cADPR-induced Ca
2+
release, also suggesting such an
interaction [33].
However, the exact molecular mechanisms under-
lying this interaction are poorly studied. In the first
study to identify a cADPR receptor, [
32
P]8-N
3
-cADPR
was used to covalently label putative cADPR binding
proteins (cADPR-BP) in sea urchin eggs [34]. As pro-
teins of 100 and 140 kDa were labelled, it was conclu-
ded that either proteolytic fragments of RyR were

labelled or that a distinct cADPR-BP mediated the
effects at the RyR (Fig. 2). A putative direct binding
site of cADPR at the RyR has not been described so
far. In contrast, in a limited number of cell systems
FK506 binding protein 12.6 (FKBP 12.6, calstabin2)
was found to bind cADPR and to mediate responsive-
ness of RyR towards cADPR [35,36]. The data sup-
port a model in which binding of FKBP 12.6 to RyR
decreases its open probability, whereas binding of
cADPR or FK506 to FKBP 12.6 weakens the inter-
action between FKBP 12.6 and RyR, thereby resulting
in an increased open probability of RyR.
Other studies have shown that in specific cell sys-
tems additional proteins must be present, e.g. that cal-
modulin effectively decreases the EC
50
for cADPR in
sea urchin egg homogenates [37] or that Tyr phos-
phorylation of the RyR enhances its responsiveness to
cADPR [38].
Ca
2+
entry by cADPR
In addition to Ca
2+
release via RyR, cADPR has been
demonstrated to activate Ca
2+
entry [18,39,40]. Ini-
tially, it was shown that microinjection of cADPR into

Jurkat T-cells induced long-lasting trains of Ca
2+
spikes that were blocked by addition of Zn
2+
or
SKF96365 [39]. Preincubation with the specific cADPR
antagonist 7-deaza-8-Br-cADPR abolished long-lasting
Ca
2+
signalling evoked by T-cell receptor ⁄ CD3 ligation
[18]. Evidence for cADPR involvement in calcium entry
was also obtained in neutrophils [40]. The chemotatic
Second messenger function of cADPR A. H. Guse
4592 FEBS Journal 272 (2005) 4590–4597 ª 2005 FEBS
peptide fMLP induced biphasic calcium signalling –
calcium release followed by calcium entry – in neu-
trophils from wild type mice. The calcium entry phase
was blocked by 8-Br-cADPR, a cADPR antagonist.
Furthermore, fMLP did not elicit the calcium entry
response in neutrophils from Cd38
– ⁄ –
mice. The
Cd38
– ⁄ –
neutrophils lack the ability to produce cADPR
[40]. These data suggest that cADPR, in addition to
Ca
2+
release, also promotes Ca
2+

entry.
What are the underlying mechanisms? A mechan-
ism generally assumed to play a role in nonexcitable
cells is the capacitative Ca
2+
entry mechanism
[reviewed in 7,41,42]. Ca
2+
currents with very low
amplitude activated by store-depletion have been
detected in several nonexcitable cells types [43,44].
Evidence for activation of store-operated Ca
2+
entry
secondary to cADPR-mediated Ca
2+
release (Fig. 2)
was obtained in RyR knock-down Jurkat T-cells in
which the long-lasting phase of Ca
2+
signalling was
partially reduced in amplitude [33]. Moreover, appli-
cation of cADPR into InsP
3
receptor-deficient DT40
cells evoked CRAC-like plasma membrane currents
[45]. Extracellular addition of the novel membrane-
permeant cADPR agonists N1-ethoxymethyl-cIDPR
(cIDPRE) and N1-[(phosphoryl-O-ethoxy)-methyl]-
N9-[(phosphoryl-O-ethoxy)-methyl]-hypoxanthine-cyclic

pyrophosphate (cIDP-DE) to intact T-cells employing
aCa
2+
-free ⁄ Ca
2+
-reintroduction protocol also sug-
gests capacitative Ca
2+
entry secondary to Ca
2+
release evoked by cADPR [46,47]. In recent years, the
plasma membrane ion channel transient receptor
potential – melastatin-like (TRPM2) has gained atten-
tion because it is activated by adenosine diphospho-
ribose (ADPR), which is synthesized from NAD by
CD38-type ADPRC and which is also a breakdown
product of cADPR (Fig. 2). TRPM2 is a Ca
2+
- and
Na
+
-permeable cation channel that is mainly
expressed in the brain and in cells of the immune sys-
tem [48–50]. The nudix box in the cytosolic C-ter-
minal region of TRPM2, a conserved motif of
enzymes with nucleotide pyrophosphatase activity,
appears to bind ADPR and regulate TRPM2
[48,49,51]. Very recently, it was shown that cADPR
can also activate TRPM2 [52]. Activation of TRPM2
by cADPR alone resulted in very small currents and

was observed only at very high cADPR concentra-
tions (EC
50
¼ 700 lm; [52]); such concentrations
likely are not present in cells, as determination of
cADPR usually resulted in low micromolar concen-
trations (e.g. [18]). Most interestingly, a likely physio-
logical concentration of 10 lm cADPR shifted the
EC
50
for ADPR from 12 lm to 90 nm [52]. Thus,
cADPR appears to be a potent coregulator for Ca
2+
(and Na
+
) entry via TRPM2. The situation, however,
appears to be more complex because physiological
concentrations of AMP inhibit the effect of ADPR
on TRPM2 channel gating. The individual contribu-
tion of each of these nucleotides to the regulation of
Ca
2+
entry under physiological conditions, e.g. with-
out washout of endogenous intracellular compounds,
will require further investigation in the future.
Structure–activity relationship of
cADPR
In-depth reviews covering the chemistry and biological
activity of many cADPR analogues have been pub-
lished [16,53–55] and the reader interested in more

complete coverage of the subject may refer to these
review articles. However, I will focus on an interesting
series of agonistic cADPR analogues recently devel-
oped. When analysing the Ca
2+
-mobilizing properties
of derivatives modified in the northern ribose of
cADPR in permeabilized T-cells, it was observed that
replacement of the hydroxyl group at C2¢¢ [for clarity
atoms of the ‘northern ribose’ will be marked as dou-
ble prime (¢¢) while atoms in the southern ribose will
be marked as single prime (¢)] by an amino group was
almost without effect on the EC
50
of Ca
2+
release
(Fig. 3; [56]). This indicates that at this side of the
molecule either the polar interactions with its interact-
ing protein were fully replaced by the amino group or
that no or only minor ligand protein interactions took
place. Astonishingly, another modification of the nor-
thern ribose, cyclic ADP carbocyclic ribose (cADPcR;
Fig. 3), showed weaker Ca
2+
release activity indicating
that the oxygen atom of the northern ribose is indeed
important for Ca
2+
release [56]. This situation is

unique for the northern ribose since replacement of the
oxygen by a carbocyclic bridge in the southern ribose
in the molecule termed cyclic aristeromycin diphos-
phoribose (cArisDPR, Fig. 3; [57]) did not significantly
alter its Ca
2+
releasing potential in permeabilized
T-cells [56]. These data indicate that both ribose moie-
ties might be suitable targets for additional and more
radical modifications.
Besides modifications in the ribose moieties, novel
analogues modified in the nucleobase show that the
base hypoxanthine can replace adenine without loss
of biological activity [58]. This is true for N1-cID-
PR, a cyclic molecule in which the cyclic bond is
made between the anomeric C1 of the northern
ribose and N1 of inosine (Fig. 4), while N7-cIDPR
showed no Ca
2+
release activity in sea urchin egg
homogenates [59]. Indeed, Ca
2+
release activity of
inosine derivatives was first described for a series of
A. H. Guse Second messenger function of cADPR
FEBS Journal 272 (2005) 4590–4597 ª 2005 FEBS 4593
cIDPR analogues that were cyclized between N1 of
inosine and C2 of the northern ribose [60]. In addi-
tion, 8-Br-N1-cIDPR induced Ca
2+

signalling in
intact T-cells [61,62]. This finding was surprising
since 8-Br-N1-cADPR is a well-known antagonist of
cADPR [63].
Fig. 3. Ca
2+
-releasing activity of some
southern and northern ribose modified
cADPR analogues.
Fig. 4. Ca
2+
-releasing activity of some cIDPR analogues.
Second messenger function of cADPR A. H. Guse
4594 FEBS Journal 272 (2005) 4590–4597 ª 2005 FEBS
A combination of nucleobase and ribose modifica-
tions led to the development of an N1-ethoxymethyl-
cIDPR (cIDPRE) in which the northern ribose was
replaced by an ether strand mimicking the C1-O-
C4 ⁄ C5 part of the original ribose (Fig. 4; [46]). Despite
this enormous modification the compound was a par-
tial agonist in permeabilized T-cells and induced both
local and global Ca
2+
signalling in intact T-cells [46].
8-Azido- and 8-NH
2
-cIDPRE performed similarly
(Fig. 4) whereas the halogenated compounds 8-Br- and
8-Cl-cIDPRE were almost without effect (Fig. 4; [46]).
An even stronger modification of the original molecule

cADPR was achieved by substitution of both the nor-
thern and southern ribose by ether strands, resulting
in N1-[(phosphoryl-O-ethoxy)-methyl]-N9-[(phosphoryl-
O-ethoxy)-methyl]-hypoxanthine-cyclic pyrophosphate
(cIDP-DE; Fig. 4). This compound was a partial agon-
ist in permeabilized cells and, when applied extracellu-
larly to intact T-cells, induced biphasic Ca
2+
signalling
comparable to T-cell receptor⁄ CD3 stimulation [47].
The biological activity of cIDP-DE is not restricted to
T-lymphocytes; extracellular addition to intact mouse
cardiac myocytes revealed activation of subcellular
Ca
2+
signalling and the induction of global Ca
2+
waves, which occurred in an oscillatory manner [47].
In terms of structure–activity relationship these data
indicate that the northern and southern riboses are pri-
marily necessary as linkers between the base adenine
(or hypoxanthine) and the diphospho-bridge, as they
can be replaced by much simpler ether strands. These
ether strands mimic the distance between the nucleo-
base and the diphospho-bridge, but on the other hand
likely are involved in polar interactions with the
cADPR receptor protein. Certainly, the natural lin-
kers, the northern and southern ribose moieties, do a
better job, as can be seen from the quantitative com-
parison with cADPR [47], probably by allowing more

interactions, but the new analogues open the possibil-
ity for the development of further, perhaps even more
simple compounds with biological activity. Such com-
pounds might be more suitable for pharmaceutical
applications as compared to the cADPR analogues
available so far.
Conclusion
Although the molecular mechanism of receptor-medi-
ated formation of cADPR is still mysterious in many
aspects, significant advancements were achieved by
demonstrating that the topological paradox of extracel-
lular ⁄ intravesicular CD38 can be circumvented by spe-
cific transport processes of the substrate NAD and the
second messenger cADPR. In addition, the description
of novel, non-CD38-like ADPRC may be a good start-
ing point for their identification in the near future. The
use of novel inosine-based cyclic nucleotides signifi-
cantly added to our understanding of the structure–
activity relationship of cADPR. Finally, a potential
new mechanism underlying Ca
2+
entry mediated by
cADPR may, in addition to capacitative Ca
2+
entry,
involve gating of TRPM2 in conjunction with ADPR.
Acknowledgements
I am grateful to my coworkers and collaboration part-
ners for their continuous support. Thanks are also
expressed to Tim Walseth (Minneapolis, USA) for crit-

ically reading the manuscript. Research in my lab is
supported by grants from the Deutsche Forschungs-
gemeinschaft (no. GU 360 ⁄ 7-3 ⁄ 9-1 ⁄ 9-2/10-1), the
Hertie-Foundation (no. 1.01.1 ⁄ 04 ⁄ 010, jointly with
Alexander Flu
¨
gel, Martinsried, Germany), the Well-
come Trust (research collaboration grant no. 068065
jointly with Barry Potter, Bath, UK) and the Deutsche
Akademische Austauschdienst (no. 423 ⁄ vrc-PPP-sr,
jointly with Li-he Zhang, Beijing, China).
References
1 Clapper DL, Walseth TF, Dargie PJ and Lee HC
(1987) Pyridine nucleotide metabolites stimulate calcium
release from sea urchin egg microsomes desensitized to
inositol trisphosphate. J Biol Chem 262, 9561–9568.
2 Lee HC, Walseth TF, Bratt GT, Hayes RN & Clapper
DL (1989) Structural determination of a cyclic metabo-
lite of NAD
+
with intracellular Ca
2+
-mobilizing activ-
ity. J Biol Chem 264, 1608–1615.
3 Kim H, Jacobson EL & Jacobson MK (1993) Position
of cyclization in cyclic ADP-ribose. Biochem Biophys
Res Commun 194, 1143–1147.
4 Lee HC, Aarhus R & Levitt D (1994) The crystal struc-
ture of cyclic ADP-ribose. Nat Struct Biol 1, 143–144.
4a Yamasaki M, Churchill GC & Galione A (2005) Cal-

cium signalling by nicotinic acid adenine dinucleotide
phosphate (NAADP). FEBS J 272, 4598–4606.
5 Strehler EE & Treiman M (2004) Calcium pumps of
plasma membrane and cell interior. Curr Mol Medical
4, 323–335.
6 Carafoli E (2005) Calcium – a universal carrier of biolo-
gical signals. FEBS J 272, 1073–1089.
7 Putney JW Jr, Broad LM, Braun FJ, Lievremont JP &
Bird GS (2001) Mechanisms of capacitative calcium
entry. J Cell Sci 114, 2223–2229.
8 Berridge MJ, Lipp P & Bootman MD (2000) The versa-
tility and universality of calcium signalling. Nat Rev
Mol Cell Biol 1, 11–21.
A. H. Guse Second messenger function of cADPR
FEBS Journal 272 (2005) 4590–4597 ª 2005 FEBS 4595
9 Schulz I & Krause E (2004) Inositol 1,4,5-trisphosphate
and its coplayers in the concert of Ca
2+
signalling –
new faces in the line up. Curr Mol Medical 4, 313–322.
10 Lee HC (2004) Multiplicity of Ca
2+
messengers and
Ca
2+
stores: a perspective from cyclic ADP-ribose and
NAADP. Curr Mol Medical 4, 227–237.
11 Galione A & Churchill GC (2002) Interactions between
calcium release pathways: multiple messengers and mul-
tiple stores. Cell Calcium 32, 343–354.

12 Bai N, Lee HC & Laher I (2005) Emerging role of cyclic
ADP-ribose (cADPR) in smooth muscle. Pharmacol
Ther 105, 189–207.
13 Gerasimenko O & Gerasimenko J (2004) New aspects
of nuclear calcium signalling. J Cell Sci 117, 3087–3094.
14 De Flora A, Zocchi E, Guida L, Franco L & Bruzzone
S (2004) Autocrine and paracrine calcium signalling by
the CD38 ⁄ NAD
+
⁄ Cyclic ADP-Ribose System. Ann NY
Acad Sci 1028, 176–191.
15 Guse AH (2004) Regulation of calcium signalling by the
second messenger cyclic adenosine diphosphoribose
(cADPR). Curr Mol Medical 4, 239–248.
16 Guse AH (2004) Biochemistry, biology, and pharmacol-
ogy of cyclic adenosine diphosphoribose (cADPR). Curr
Medical Chem 11, 847–855.
17 Higashida H (1997) ADP-ribosyl cyclase coupled with
receptors via G proteins. FEBS Lett 418, 355–356.
18 Guse AH, da Silva CP, Berg I, Skapenko AL, Weber
K, Heyer P et al. (1999) Regulation of calcium signal-
ling in T lymphocytes by the second messenger cyclic
ADP-ribose. Nature 398, 70–73.
19 Hellmich MR & Strumwasser F (1991) Purification and
characterization of a molluscan egg-specific NADase, a
second-messenger enzyme. Cell Regul 2, 193–202.
20 Glick DL, Hellmich MR, Beushausen S, Tempst P,
Bayley H & Strumwasser F (1991) Primary structure of
a molluscan egg-specific NADase, a second-messenger
enzyme. Cell Regul 2, 211–218.

21 Schuber F & Lund FE (2004) Structure and enzymology
of ADP-ribosyl cyclases: conserved enzymes that pro-
duce multiple calcium mobilizing metabolites. Curr Mol
Medical 4, 249–261.
22 De Flora A, Franco L, Guida L, Bruzzone S, Usai C &
Zocchi E (2000) Topology of CD38. Chem Immunol 75,
79–98.
23 Bruzzone S, Guida L, Zocchi E, Franco L & De Flora
A (2001) Connexin 43 hemi channels mediate Ca
2+
-
regulated transmembrane NAD
+
fluxes in intacT-cells.
FASEB J 15, 10–12.
24 Guida L, Bruzzone S, Sturla L, Franco L, Zocchi E &
De Flora A (2002) Equilibrative and concentrative
nucleoside transporters mediate influx of extracellular
cyclic ADP-ribose into 3T3 murine fibroblasts. J Biol
Chem 277, 47097–47105.
25 Bruzzone S, Franco L, Guida L, Zocchi E, Contini P,
Bisso A, Usai C & De Flora A (2001) A self-restricted
CD38-connexin 43 cross-talk affects NAD
+
and cyclic
ADP-ribose metabolism and regulates intracellular cal-
cium in 3T3 fibroblasts. J Biol Chem 276, 48300–48308.
26 Podesta M, Benvenuto F, Pitto A, Figari O, Bacigalupo
A, Bruzzone S et al. (2005) Concentrative uptake of cyc-
lic ADP-ribose generated by BST-1

+
stroma stimulates
proliferation of human hematopoietic progenitors.
J Biol Chem 280, 5343–5349.
27 Graeff RM, Franco L, De Flora A & Lee HC (1998)
Cyclic GMP-dependent and -independent effects on the
synthesis of the calcium messengers cyclic ADP-ribose
and nicotinic acid adenine dinucleotide phosphate.
J Biol Chem 273, 118–125.
28 Matsumura N & Tanuma S (1998) Involvement of cyto-
solic NAD
+
glycohydrolase in cyclic ADP-ribose meta-
bolism. Biochem Biophys Res Commun 253, 246–252.
29 Bruzzone S, De Flora A, Usai C, Graeff R & Lee HC
(2003) Cyclic ADP-ribose is a second messenger in the
lipopolysaccharide-stimulated proliferation of human
peripheral blood mononuclear cells. Biochem J 375,
395–403.
30 Sternfeld L, Krause E, Guse AH & Schulz I (2003)
Hormonal control of ADP-ribosyl cyclase activity in
pancreatic acinar cells from rats. J Biol Chem 278,
33629–33636.
31 Ceni C, Muller-Steffner H, Lund F, Pochon N, Schweit-
zer A, De Waard M et al. (2003) Evidence for an intra-
cellular ADP-ribosyl cyclase ⁄ NAD
+
-glycohydrolase in
brain from CD38-deficient mice. J Biol Chem 278,
40670–40678.

32 Galione A, Lee HC & Busa WB (1991) Ca
2+
-induced
Ca
2+
release in sea urchin egg homogenates: modula-
tion by cyclic ADP-ribose. Science 253, 1143–1146.
33 Schwarzmann N, Kunerth S, Weber K, Mayr GW &
Guse AH (2002) Knock-down of the type 3 ryanodine
receptor impairs sustained Ca
2+
signalling via the T-cell
receptor ⁄ CD3 complex. J Biol Chem 277, 50636–50642.
34 Walseth TF, Aarhus R, Kerr JA & Lee HC (1993) Iden-
tification of cyclic ADP-ribose-binding proteins by
photoaffinity labeling. J Biol Chem 268 , 26686–26691.
35 Noguchi N, Takasawa S, Nata K, Tohgo A, Kato I,
Ikehata F et al. (1997) Cyclic ADP-ribose binds to
FK506-binding protein 12.6 to release Ca
2+
from islet
microsomes. J Biol Chem 272, 3133–3136.
36 Tang WX, Chen YF, Zou AP, Campbell WB & Li PL
(2002) Role of FKBP12.6 in cADPR-induced activation
of reconstituted ryanodine receptors from arterial
smooth muscle. Am J Physiol Heart Circ Physiol 282,
H1304–1310.
37 Lee HC, Aarhus R, Graeff R, Gurnack ME & Walseth
TF (1994) Cyclic ADP ribose activation of the ryano-
dine receptor is mediated by calmodulin. Nature 370,

307–309.
38 Guse AH, Tsygankov AY, Weber K & Mayr GW
(2001) Transient tyrosine phosphorylation of human
Second messenger function of cADPR A. H. Guse
4596 FEBS Journal 272 (2005) 4590–4597 ª 2005 FEBS
ryanodine receptor upon T-cell stimulation. J Biol Chem
276, 34722–34727.
39 Guse AH, Berg I, da Silva CP, Potter BV & Mayr GW
(1997) Ca
2+
entry induced by cyclic ADP-ribose in
intact T-lymphocytes. J Biol Chem 272, 8546–85450.
40 Partida-Sanchez S, Cockayne DA, Monard S, Jacobson
EL, Oppenheimer N, Garvy B et al. (2001) Cyclic ADP-
ribose production by CD38 regulates intracellular cal-
cium release, extracellular calcium influx and chemotaxis
in neutrophils and is required for bacterial clearance
in vivo. Nat Medical 7, 1209–1216.
41 Putney JW Jr (1986) A model for receptor-regulated cal-
cium entry. Cell Calcium 7, 1–12.
42 Bird GS, Aziz O, Lievremont JP, Wedel BJ, Trebak M,
Vazquez G & Putney JW Jr (2004) Mechanisms of
phospholipase C-regulated calcium entry. Curr Mol
Medical 4, 291–301.
43 Hoth M & Penner R (1992) Depletion of intracellular
calcium stores activates a calcium current in masT-cells.
Nature 355, 353–356.
44 Hoth M, Fasolato C & Penner R (1993) Ion channels
and calcium signalling in mast-cells. NY Acad Sci 707,
198–209.

45 Kiselyov K, Shin DM, Shcheynikov N, Kurosaki T &
Muallem S (2001) Regulation of Ca
2+
-release-activated
Ca
2+
current (ICRAC) by ryanodine receptors in ino-
sitol 1,4,5-trisphosphate-receptor-deficient DT40 cells.
Biochem J 360, 17–22.
46 Gu X, Yang Z, Zhang L, Kunerth S, Fliegert R, Weber
K et al. (2004) Synthesis and biological evaluation of
novel membrane-permeant cyclic ADP-ribose mimics:
N1-[(5¢¢-O-phosphorylethoxy)methyl]-5¢-O-phosphoryl-
inosine 5¢,5¢¢-cyclicpyrophosphate (cIDPRE) and 8-sub-
stituted derivatives. J Medical Chem 47, 5674–5682.
47 Guse AH, Gu X, Zhang L, Weber K, Kramer E, Yang
Z et al. (2005) A minimal structural analogue of cyclic
ADP-ribose: Synthesis and calcium release activity in
mammalian cells. J Biol Chem 280 , 15952–15959.
48 Perraud AL, Fleig A, Dunn CA, Bagley LA, Launay P,
Schmitz C et al. (2001) ADP-ribose gating of the cal-
cium-permeable LTRPC2 channel revealed by Nudix
motif homology. Nature 411, 595–599.
49 Sano Y, Inamura K, Miyake A, Mochizuki S, Yokoi H,
Matsushime H & Furuichi K (2001) Immunocyte Ca
2+
influx system mediated by LTRPC2. Science 293, 1327–
1330.
50 Perraud AL, Shen B, Dunn CA, Rippe K, Smith MK,
Bessman MJ et al. (2003) NUDT9, a member of the

Nudix hydrolase family, is an evolutionarily conserved
mitochondrial ADP-ribose pyrophosphatase. J Biol
Chem 278, 1794–1801.
51 Kuhn FJ & Luckhoff A (2004) Sites of the NUDT9-H
domain critical for ADP-ribose activation of the cation
channel TRPM2. J Biol Chem 279, 46431–46437.
52 Kolisek M, Beck A, Fleig A & Penner R (2005) Cyclic
ADP-ribose and hydrogen peroxide synergize with
ADP-ribose in the activation of TRPM2 channels. Mol
Cell 18, 61–69.
53 Zhang FJ, Gu QM & Sih CJ (1999) Bioorganic chemis-
try of cyclic ADP-ribose (cADPR). Bioorg Medical
Chem 7, 653–664.
54 Shuto S & Matsuda A (2004) Chemistry of cyclic ADP-
ribose and its analogs. Curr Medical Chem 11, 827–845.
55 Potter BV & Walseth TF (2004) Medicinal chemistry
and pharmacology of cyclic ADP-ribose. Curr Mol
Medical 4, 303–311.
56 Guse AH, Cakir-Kiefer C, Fukuoka M, Shuto S, Weber
K, Bailey VC et al. (2002) Novel hydrolysis-resistant
analogues of cyclic ADP-ribose: modification of the
‘northern’ ribose and calcium release activity. Biochemis-
try 41, 6744–6751.
57 Bailey VC, Fortt SM, Summerhill RJ, Galione A & Pot-
ter BV (1996) Cyclic aristeromycin diphosphate ribose:
a potent and poorly hydrolysable Ca
2+
-mobilizing
mimic of cyclic adenosine diphosphate ribose. FEBS
Lett 379, 227–230.

58 Wagner GK, Guse AH & Potter BVL (2005) Rapid syn-
thetic route towards structurally modified derivatives of
cyclic adenosine 5¢-diphosphate ribose. J Org Chem 70,
4810–4819.
59 Graeff RM, Walseth TF, Hill HK, Lee HC (1996)
Fluorescent analogs of cyclic ADP-ribose: synthesis,
spectral characterization, and use. Biochemistry 35,
379–386.
60 Huang LJ, Zhao YY, Yuan L, Min JM & Zhang LH
(2002) Syntheses and calcium-mobilizing evaluations of
N1-glycosyl-substituted stable mimics of cyclic ADP-
ribose. J Medical Chem 45, 5340–5352.
61 Wagner GK, Black S, Guse AH & Potter BV (2003)
First enzymatic synthesis of an N1-cyclised cADPR
(cyclic-ADP ribose) analogue with a hypoxanthine par-
tial structure: discovery of a membrane permeant
cADPR agonist. Chem Commun (Camb) 15, 1944–1945.
62 Wagner GK, Riley AM, Rosenberg HJ, Taylor CW,
Guse AH & Potter BV (2003) Analogues of cyclic ade-
nosine 5¢-diphosphate ribose and adenophostin A,
nucleotides in cellular signal transduction. Nucleic Acids
Res Suppl 3, 1–2.
63 Walseth TF & Lee HC (1993) Synthesis and characteri-
zation of antagonists of cyclic-ADP-ribose-induced
Ca
2+
release. Biochim Biophys Acta 1178, 235–242.
A. H. Guse Second messenger function of cADPR
FEBS Journal 272 (2005) 4590–4597 ª 2005 FEBS 4597

×