13
Journal of the Iranian Chemical Society, Vol. 2, No. 3, September 2005, pp. 176-188. JOURNAL OF THE Iranian Chemical Society GPCRs as Therapeutic Targets: a View on Adenosine Receptors Structure and Functions, and Molecular Modeling Support D. Dal Ben, C. Lambertucci, S. Vittori, R. Volpini and G. Cristalli* Dipartimento di Scienze Chimiche,Via S. Agostino, 1; Università di Camerino, 62032 Camerino, Italy (Received 30 July 2005, Accepted 8 August 2005) G-protein-coupled receptors (GPCRs) represent the largest known family of signal-transducing proteins and transmit signals for light and many extracellular regulatory molecules. GPCRs are dysfunctional or dysregulated in several human diseases and are estimated to be the targets of ~40% of the drugs used in clinical medicine today. Receptors for adenosine belong to this family, and so far four subtypes, the A 1 , A 2A , A 2B , and A 3 , have been recognized. The activation of adenosine receptors (ARs) is largely responsible for the variety of effects produced by adenosine throughout several organ systems. Based on the wide (and often beneficial) effects attributed to the accumulation of endogenously released adenosine, it has long been considered that regulation of ARs has considerable therapeutic potential. In this review, we focus on recent work on adenosine receptors as therapeutic targets and, in particular, on molecular modelling support to adenosine receptors targeting. Keywords: G-protein-coupled receptors, Signal transduction, Adenosine, Adenosine receptors, Homology modeling INTRODUCTION G-protein-coupled receptors (GPCRs) compose a big set of sensory proteins that mediate a wide variety of physiological processes by responding to different external stimuli such as hormones, paracrines (local hormones), neurotransmitters, neuromodulators, odours, and light [1-3]. These transmembrane receptors transduce the external signals mostly by interacting with regulatory G-proteins [4-6]. Such proteins transmit the signal to effector enzymes, such as cytoplasmic proteins and ion channels, by causing rapid changes in the concentration of intracellular signalling molecules, cAMP, cGMP, diacylglycerol, arachidonic acid, inositol phosphates, cytosolic ions. As some GPCRs may use heterotrimeric G- proteins and even other cytoplasmic enzymes as transducers in * Corresponding author. E-mail: [email protected] their signalling and others may interact with non-G-protein transducers only, some authors prefer alternative names for this protein superfamily such as 7-transmembrane receptors, heptahelical receptors, or serpentine-like receptors [7,8]. Although the GPCR families do not share sequence similarity with each other, specific features exist in all GPCR classes [9]. For example, all these receptors have in common a central core domain that consists of seven transmembrane (TM1-7) helices connected by three intracellular loops (IL1, IL2, and IL3) and three extracellular loops (EL1, EL2, and EL3). Two cysteine residues, one in TM3/EL1 interface and one in EL2, which are conserved in almost all GPCRs, form an essential disulfide link responsible for the packing and stabilization of a restricted number of conformations of these seven TM domains (Fig. 1). Beside sequence variations, GPCRs differ in the length and function of their N-terminal extracellular domain, their C-terminal intracellular domain and their intracellular loops. Each of these domains confers

GPCRs as therapeutic targets: a view on adenosine receptors structure and functions, and molecular modeling support

Embed Size (px)

Citation preview

Journal of the Iranian Chemical Society, Vol. 2, No. 3, September 2005, pp. 176-188.

JOURNAL OF THE Iranian Chemical Society

GPCRs as Therapeutic Targets: a View on Adenosine Receptors Structure and Functions, and Molecular Modeling Support

D. Dal Ben, C. Lambertucci, S. Vittori, R. Volpini and G. Cristalli* Dipartimento di Scienze Chimiche,Via S. Agostino, 1; Università di Camerino, 62032 Camerino, Italy

(Received 30 July 2005, Accepted 8 August 2005)

G-protein-coupled receptors (GPCRs) represent the largest known family of signal-transducing proteins and transmit signals

for light and many extracellular regulatory molecules. GPCRs are dysfunctional or dysregulated in several human diseases and are

estimated to be the targets of ~40% of the drugs used in clinical medicine today. Receptors for adenosine belong to this family,

and so far four subtypes, the A1, A2A, A2B, and A3, have been recognized. The activation of adenosine receptors (ARs) is largely

responsible for the variety of effects produced by adenosine throughout several organ systems. Based on the wide (and often

beneficial) effects attributed to the accumulation of endogenously released adenosine, it has long been considered that regulation

of ARs has considerable therapeutic potential. In this review, we focus on recent work on adenosine receptors as therapeutic

targets and, in particular, on molecular modelling support to adenosine receptors targeting.

Keywords: G-protein-coupled receptors, Signal transduction, Adenosine, Adenosine receptors, Homology modeling

INTRODUCTION

G-protein-coupled receptors (GPCRs) compose a big set of

sensory proteins that mediate a wide variety of physiological

processes by responding to different external stimuli such as

hormones, paracrines (local hormones), neurotransmitters,

neuromodulators, odours, and light [1-3]. These

transmembrane receptors transduce the external signals mostly

by interacting with regulatory G-proteins [4-6]. Such proteins

transmit the signal to effector enzymes, such as cytoplasmic

proteins and ion channels, by causing rapid changes in the

concentration of intracellular signalling molecules, cAMP,

cGMP, diacylglycerol, arachidonic acid, inositol phosphates,

cytosolic ions. As some GPCRs may use heterotrimeric G-

proteins and even other cytoplasmic enzymes as transducers in

* Corresponding author.

E-mail: [email protected]

their signalling and others may interact with non-G-protein

transducers only, some authors prefer alternative names for

this protein superfamily such as 7-transmembrane receptors,

heptahelical receptors, or serpentine-like receptors [7,8].

Although the GPCR families do not share sequence

similarity with each other, specific features exist in all GPCR

classes [9]. For example, all these receptors have in common a

central core domain that consists of seven transmembrane

(TM1-7) helices connected by three intracellular loops (IL1,

IL2, and IL3) and three extracellular loops (EL1, EL2, and

EL3). Two cysteine residues, one in TM3/EL1 interface and

one in EL2, which are conserved in almost all GPCRs, form

an essential disulfide link responsible for the packing and

stabilization of a restricted number of conformations of these

seven TM domains (Fig. 1). Beside sequence variations,

GPCRs differ in the length and function of their N-terminal

extracellular domain, their C-terminal intracellular domain and

their intracellular loops. Each of these domains confers

Dal Ben et al.

177

specific properties to these receptor proteins.

The superfamily of GPCRs can be subdivided into seven

families of receptors whose protein sequences share

significant similarity [10]. The main family (Family A), which

comprises rhodopsin and adrenoceptors, is composed by the

majority of GPCRs identified to date, and is the best studied

GPCR family both structurally and functionally. Receptors

that belong to this family are activated by a variety of stimuli,

including photons, odorants, and hormones and

neurotransmitters with molecular structures ranging from

small biogenic amines (e.g., catecholamines) to peptides (e.g.,

gonadotropin-releasing hormone) and complex glycoproteins

(e.g., luteinizing hormone). The other subfamilies are the

secretin-vasointestinal peptide (VIP) family (Family B),

members of which bind neuropeptides and peptide hormones,

the metabotropic glutamate receptor family (Family C),

comprising at least eight strongly related subtypes of

glutamate binding receptors, the fungal pheromone P family

(Family D), the fungal pheromone A receptors (Family E), and

the cAMP receptors of dictyostelium discoideum (Family F).

GPCRs can exist as dimers or as part of larger oligomeric

complexes, and many studies have intimated the existence of

heterodimeric GPCR pairings. Determination of the

prevalence and relevance of these complexes in native tissues

and the implications of heterodimerization for pharmacology

and potentially for drug design is still a key issue [11,12].

The superfamily of GPCRs comprises receptors for many

substances with important physiological functions. For this

reason their dysfunction leads to human diseases, and in fact

many GPCRs are targets for pharmaceuticals and drugs of

abuse [13-15]. It has been estimated that ~80% of known

hormones and neurotransmitters activate cellular signal

transduction mechanisms by activating GPCRs, and it has

been estimated that GPCRs represent ~40% of current drug

targets.

ADENOSINE RECEPTORS

Adenosine The purine nucleoside adenosine (Fig. 2) is a mediator

playing an important role in modulating many

(patho)physiological conditions in the CNS as well as in

peripheral organs and tissues [16,17].

Adenosine is ubiquitously released in the extracellular

space from metabolically active cells and is generated by the

degradation of released ATP. As adenosine is unstable and has

a short half-life because of deamination or cellular reuptake,

hypoxia-induced increases typically bear only on local

adenosine receptor signalling. Hence, adenosine fulfils the

criteria of an autacoid. In the nervous system adenosine is

“omnipresent”, i.e. exists in all cells, and is released from

apparently all cells, including neurones and glia [18,19].

While ATP may function as a neurotransmitter in some brain

areas, adenosine is neither stored nor released as a classical

neurotransmitter since it does not accumulate in synaptic

vesicles, being released from the cytoplasm into the

extracellular space through a nucleoside transporter. The

adenosine transporters also mediate adenosine reuptake, and

the direction of the transport is dependent upon the

concentration gradient at both sides of the membrane. There

are two sources of extracellular adenosine: release of

adenosine by facilitated diffusion and extracellular conversion

of released adenine nucleotides into adenosine through a series

of ectoenzymes. Interestingly, extracellular adenosine levels

may be under control of adenosine A1 receptor since blockade

of A1 receptors increases the extracellular concentration of

adenosine in cardiac fibroblasts [20]. However, as the

extracellular formation of adenosine from released adenine

nucleotides represents a second source of adenosine, which is

not affected by transport inhibition, the transport inhibitors

usually cause an increase in the extracellular adenosine levels

[21].

Adenosine extracellular concentration is calculated to be in

the range of 50-200 nM [22], but under several adverse

conditions, including ischemia, trauma, stress and

inflammation, extracellular levels of adenosine are increased

due to increased energy demands and ATP metabolism. Under

hypoxic and ischaemic conditions there is a marked increase

in cytoplasmatic adenosine leading to an intense release of

adenosine, which is inhibited by adenosine uptake inhibitors

[16]. In the isolated heart hypoxia raises the extracellular

concentration by more than 20-fold, reaching concentrations

of > 1 μM [23].

GPCRs as Therapeutic Targets

178

Fig. 1. General architecture of GPCRs.

Adenosine Receptors Adenosine receptors (ARs) are members of the

superfamily of G-protein-coupled receptors (GPCRs), with

four subtypes currently recognized, the A1, A2A, A2B, and A3.

Adenosine receptors (ARs) are present on virtually every

tissue and are often co-expressed in the same cell type, and

their activation is predominantly responsible for the wide

variety of effects produced by adenosine throughout several

organ systems [17]. All four subtypes of adenosine receptors

are encoded by distinct genes, and their expression is

widespread such that adenosine controls the function of

virtually every organ and tissue. However, receptor subtype

distribution and densities vary greatly. With the exception of

the A3, the existence of AR subtypes in various tissues had

been appreciated prior to their cloning as a result of

pharmacological characterization. The cloning of the four AR

subtypes has allowed for significant progresses to be made in

the understanding of several aspects of AR activity at a

molecular level.

N

N

NH2

N

NO

OHHO

HO

Fig. 2. Adenosine.

The large variations in the local adenosine concentrations

support the experimentally derived concept that cell surface

receptors exist with high (A1, A2A, A3) and low affinity (A2B)

for the purine nucleoside. Originally, the receptors were

classified based on their affinities for adenosine analogues and

methylxanthine antagonists [24]. Adenosine receptors can also

be differentiated according to their preferred mechanism of

signal transduction: A1 and A3 receptors interact with pertussis

toxin-sensitive G-proteins of the Gi family, while the

canonical signalling mechanism of the A2A and of the A2B

receptors is stimulation of adenylyl cyclase via Gs (Fig. 3)

[25].

Activation of A1 receptors by adenosine leads to activation

of the Go family, which leads directly to the stabilization of the

neuronal membrane potential by increasing, in particular, the

conductance of K+ ions (efflux from inside to outside the cell),

thereby counteracting excessive membrane depolarisation

[26]. A2B and A3 receptors can activate phospholipase C and

Fig. 3. Adenosine receptors and their intracellular effectors.

Dal Ben et al.

179

this is thought to be mediated by activation of Gq.

Diacylglycerol (DAG) and inositol (1,4,5)-trisphosphate (IP3)

are implicated in the regulation of protein kinase C (PKC)

activity and the intracellular concentration of Ca2+ ions,

respectively [27]. In the brain, by the inhibition of neuronal

Ca2+ influx (through inhibition of adenylyl cyclase), adenosine

counteracts the presynaptic release of the potentially

excitotoxic neurotransmitters glutamate and aspartate, which

can impair intracellular Ca2+ homeostasis through

metabotropic glutamate receptors or the induction of

membrane depolarization through ion channel-linked

glutamate receptors [28,29]. Other G-proteins can also be

activated following adenosine receptor activation even though

the physiological significance of this is unknown.

Adenosine Receptors Structure ARs display the topology typical of GPCRs. These

receptors have in common a central core domain of seven

transmembrane helices, with each TM composed of 20-25

amino acids, connected by three intracellular (IL1, IL2 and

IL3) and three extracellular (EL1, EL2 and EL3) loops. Two

cysteine residues (one in TM3/EL1 interface and one in EL2)

which are conserved in most GPCRs, form a disulfide bond

which is crucial for the folding and for the stabilization of a

limited number of conformations of these seven TMs (Fig. 4)

[17,30].

Adenosine receptors differ in the length and function of

their N-terminal extracellular domain, their C-terminal

intracellular domain and their intracellular loops. Each of

these domains provides very specific properties to these

receptor proteins. Particularly, consensus sites for N-linked

glycosylation exist on the extracellular regions of ARs,

although the precise location of the sites for this post-

translational modification varies among the AR subtypes. The

carboxyl-terminal tails of the A1, A2B, and A3, but not the A2A,

possess a conserved cysteine residue that may putatively serve

as a site for receptor palmitoylation and permit the formation

of a fourth intracellular loop. However, site-directed

mutagenesis of this residue has not been performed for any

AR subtype, and no role for putative AR palmitoylation has

been described.

The A1, A2B, and A3 receptors are very similar in regard to

the number of amino acids composing their primary structure,

Fig. 4. Schematic view of adenosine A3 receptor structure.

and overall, these AR subtypes are among the smallest

members of the GPCR family. For example, the human

homologues of A1, A2B, and A3 consist of 326, 328, and 318

amino acid residues, respectively. On the contrary, the human

A2A is composed of 409 amino acids. All cloned species

homologues of the A2A are of similar mass, and this relatively

large size is manifested in the carboxyl-terminal tail of the

receptor, which is much longer than that of the other AR

subtypes. It should be noted that the size of ARs deduced from

their primary amino acid structure frequently is not consistent

with the mass estimated by polyacrylamide gel electrophoresis

of the expressed proteins. This is probably due to the post-

translational glycosylation of ARs which may vary in a cell

type-dependent fashion. The human A1 and A3 receptors

display about 49% overall sequence identity at the amino acid

level, while the human A2A and human A2B receptors are 45%

identical (see Table 1).

Adenosine Receptors as Therapeutic Targets Research on adenosine receptors during the past 20 years

has resulted in considerable progress in identifying selective

agonists and antagonists and an increased understanding of the

particular roles adenosine receptor subtypes play in

physiological processes [31].

Indeed, several protective effects of adenosine have been

widely reported: a) adenosine has protective effects in the

GPCRs as Therapeutic Targets

180

ischemic and reperfused myocardium by minimizing the

workload of the heart and its oxygen demand during ischemia

(through adenosine A1 receptor pathways [32]) and by causing

coronary vasodilatation and increasing myocardial oxygen

supply through the activation of adenosine A2A receptors on

vascular smooth muscle and endothelial cells; beside that,

adenosine inhibits platelet aggregation, protecting endothelial

cell, integrity and limiting the vascular injury. b) Adenosine

exhibits protecting effects against cerebral ischemia. It has

been reported that stimulation of adenosine A1 receptor

decreases the mortality associated with focal or global brain

ischemia. Additionally, by acting through adenosine A2A

receptor pathways, adenosine ameliorates post-ischemic

cerebral blood flow and decreases post-ischemic cerebral

damage. c) Adenosine has been reported to be beneficial in

organ transplantation, acting by mechanisms similar to those

for its action against ischemia and reperfusion injury. d)

Adenosine is an endogenous neuroprotective agent and

exhibits anticonvulsant effects. The anticonvulsant effects of

adenosine appear to be mediated primarily by adenosine A1

receptors [33]. e) Adenosine inhibits platelet aggregation and

attenuates tissue factor expression on endothelial cells induced

by various stimuli, acting mainly through adenosine A2A

receptor pathways. For this reason, the inhibitory effects of

adenosine on platelet and endothelial cell activation are

thought to be beneficial for the treatment of thrombosis. f)

Adenosine exerts multiple effects on pain transmission both at

spinal and peripheral sites. In the spinal cord, adenosine

provides pain relief through adenosine A1 receptor pathways

in acute nociceptive, inflammatory, and neuropathic pain tests

[34]. g) Adenosine is released at sites of inflammation and acts

as an endogenous anti-inflammatory agent, and adenosine A2A

receptor pathways seem to have an important role in the anti-

inflammatory effects of adenosine. h) In asthmatic conditions,

the extracellular concentration of adenosine increases in the

airway tissue. It inhibits eosinophil or lymphocyte function

and modulates reactive oxygen species generation in

neutrophils; anyway, many aspects of this mechanisms are still

unclear [35-37].

This knowledge has continued to stimulate considerable

interest in pursuing adenosine receptors as therapeutic targets.

For example, a number of selective agonists for A1, A2A, A2B,

and A3 [38] adenosine receptors have been developed, and

several such compounds are undergoing clinical trials for the

treatment of cardiovascular diseases (A1 and A2A) [31,39-41],

pain (A1), wound healing (A2A), diabetic foot ulcers (A2A),

colorectal cancer (A3) [42] and rheumatoid arthritis (A3). In

general, adenosine receptor agonists are derivatives of the

physiological agonist adenosine.

The development of adenosine receptor agonists has been

limited by an essential requirement for retention of the ribose

moiety for agonist activity. Despite this restriction, significant

progress has been made in the identification of potent and

selective adenosine receptor agonists. On the other hand, the

clinical evaluation of some A1 and A2A adenosine receptor

agonists has been discontinued, and the major problems

comprise: side effects due to the wide distribution of

adenosine receptors; low brain penetration, which is important

for the targeting of CNS diseases; short half-lives of

compounds; lack of effects, in some cases perhaps due to

receptor desensitisation or to low receptor density in the

targeted tissue [43,44].

Partial agonists, inhibitors of adenosine metabolism

(adenosine kinase and deaminase inhibitors) or allosteric

activators of adenosine receptors may be advantageous for

certain indications, as they may exhibit fewer side effects [45].

Adenosine itself is used as a therapeutic agent for the

treatment of supraventricular paroxysmal tachycardia and

arrhythmias and, as a vasodilatatory agent, in cardiac imaging

[46,47]. Two adenosine products, Adenocard® and

Adenoscan®, have been approved by FDA and are currently

available for cardiac arrhythmias treatment and for cardiac

imaging, respectively [48,49].

Recent focus has been on the cardioprotective and

neuroprotective effects associated with AR activation during

periods of cardiac and cerebral ischemia, respectively. On the

other hand, it has been recently proposed that antagonists of

distinct AR subtypes may be used in the treatment of certain

neurological diseases such as Parkinson's disease[50-52] .

MOLECULAR MODELING SUPPORT IN ADENOSINE RECEPTORS TARGETING The 3D-structure Problem The evolution of the field of computer-aided design of ligands (both agonists and antagonists) for GPCRs, including

Dal Ben et al.

181

adenosine receptors, has depended on the availability of

suitable molecular receptor templates. In fact, due to technical

difficulties, which complicate experimental X-ray diffraction

and NMR structure determination of GPCRs, the 3D structure

of most GPCRs is still unknown [1,9]. In fact, while it is quite

simple to deduce the amino acid sequence of a protein from

the DNA sequence of the gene encoding it, determining the

three-dimensional molecular structure of proteins has proven

to be more complex, especially for membrane proteins that

represent about half of all currently exploited drug targets. The

usual time needed to solve an eukaryotic protein target from

clone to three-dimensional structure has been one to three

years for soluble proteins and even longer, with a higher risk

of failure, for membrane proteins.

Indeed, atomic resolution crystal structures of soluble

proteins have been reported in a rapidly increasing number

over the last decade, while such improvement has not been

made in terms of membrane proteins, which have proven

extremely difficult to crystallize for two main factors: the

amphipathic nature of their surface, with a hydrophobic area in

contact with membrane phospholipids and polar surface areas

in contact with the aqueous phases on both sides of the

membrane; the very low concentrations in tissues of the

majority of medically relevant membrane proteins.

Early structural data of G-protein-coupled receptors came

from low-resolution projection maps from cryo-electron

microscopy experiments. In a paper published in 1975,

Henderson and Unwin demonstrated that bacteriorhodopsin, a

protein from halobacterium halobium that works as a light-

driven proton pump, has seven membrane spanning domains

that were interpreted as α-helices [53]. The idea of a seven

transmembrane α-helical (TMH) architecture of GPCRs,

initially based on bacteriorhodopsin which does not act

through a G-protein, was well established and had already

been proposed in lots of scientific papers suggesting GPCR

models, when strong support for the 7 TMH architecture of

GPCRs came from Henderson’s group in 1993.

An electron projection map of visual rhodopsin, a

chromoprotein in the retina that acts via a G-protein,

transducin, showed seven membrane spanning domains that

appeared to be α-helices, arranged in a slightly different way

respect those in bacteriorhodopsin [54]. New suppositions

about the structure of these receptors came from

cryomicroscopic studies of rhodopsin that indicated the

existence of seven transmembrane segments and gave an

indication of the relative disposition of these TMs [55]. The

final proof of the 7 TMH architecture of rhodopsin came when

an atomic-resolution crystal structure of bovine rhodopsin was

published a few years later [56].

Adenosine Receptors Structure-based Drug Design Molecular modeling provides a useful methodological

alternative to experimental structure determination, especially

for membrane proteins. Homology modeling of a protein

structure is based on three factors: a three-dimensional

structural template, an amino acid sequence alignment of the

modelled molecule with the template molecule (and other

related molecules), and particularly adapted computer

software. The accuracy and consistency of the protein model

depends on the accuracy of the structural data template, and

the similarity of the modelled structure with the one or those

used as a template for the initial protein model. Compared to

modeling of drug targets, which usually are macromolecules

and in most cases proteins with several hundred amino acids,

modeling of drug molecules is relatively straightforward. The

challenge in modeling of drug-receptor interactions lies in

modeling of the target molecule, and in the docking of ligand

molecules into postulated binding sites.

An early model of the TM segments of an adenosine A3

receptor (cloned from rat) was proposed by van Galen [57].

This model was based on the primary sequence and structural

homology with bacteriorhodopsin, in analogy to the method

proposed by Hibert [58]. A putative ligand binding region was

proposed and explored by docking several antagonists into the

TM domain model. In particular, this model suggested that

TM3, TM6 and TM7 might be involved in ligand binding.

However, as reported above, bacteriorhodopsin lacks any

functional or sequence homology with GPCRs. The original

model of van Galen was improved in the description of

ligand/receptor interactions by introduction of a simple way to

simulate the reorganization of the native receptor structure

induced by ligand coordination (cross-docking methodology)

[59,60]. This method gives good results in predicting local

structural changes induced by ligand in the receptor binding

site. The presence of both agonist and antagonist within the

receptor can produce a simultaneous adjustment in the

GPCRs as Therapeutic Targets

182

Table 1. Sequence Alignment of Human Adenosine A1, A2A, A2B ,and A3 Receptors.

The Respective Regions Containing Transmembrane (TM) Segments Have

been Aligned with ClustalW (http://www.ebi.ac.uk/clustalw). The Residues

Color Scheme is According to ClustalW: Color Red Corresponds to Small and

Hydrophobic Residues (AVFPMILW); Color Blue is for Acidic Residues

(DE); Color Magenta is for Basic Residues (RK) and Color Green

Corresponds to the Remaining Residues (STYHCNGQ)

TM1

AA1R_HUMAN AAYIGIEVLIALVSVPGNVLVIWAVK 35

AA2AR_HUMAN SVYITVELAIAVLAILGNVLVCWAVW 32

AA2BR_HUMAN ALYVALELVIAALSVAGNVLVCAAVG 33

AA3R_HUMAN VTYITMEIFIGLCAIVGNVLVICVVK 38

TM2

AA1R_HUMAN DATFCFIVSLAVADVAVGALVIPLAI 67

AA2AR_HUMAN NVTNYFVVSLAAADIAVGVLAIPFAI 64

AA2BR_HUMAN TPTNYFLVSLAAADVAVGLFAIPFAI 65

AA3R_HUMAN TTTFYFIVSLALADIAVGVLVMPLAI 70

TM3

AA1R_HUMAN LMVACPVLILTQSSILALLAIAVDRY 106

AA2AR_HUMAN LFIACFVLVLTQSSIFSLLAIAIDRY 103

AA2BR_HUMAN LFLACFVLVLTQSSIFSLLAVAVDRY 104

AA3R_HUMAN LFMTCLLLIFTHASIMSLLAIAVDRY 109

TM4

AA1R_HUMAN RRAAVAIAGCWILSFVVGLTPMFGWN 147

AA2AR_HUMAN TRAKGIIAICWVLSFAIGLTPMLGWN 144

AA2BR_HUMAN TRARGVIAVLWVLAFGIGLTPFLGWN 145

AA3R_HUMAN RRIWLALGLCWLVSFLVGLTPMFGWN 150

TM5

AA1R_HUMAN YMVYFNFFVWVLPPLLLMVLIYLEVF 204

AA2AR_HUMAN YMVYFNFFACVLVPLLLMLGVYLRIF 201

AA2BR_HUMAN YMVYFNFFGCVLPPLLIMLVIYIKIF 206

AA3R_HUMAN YMVYFSFLTWIFIPLVVMCAIYLDIF 201

TM6

AA1R_HUMAN IAKSLALILFLFALSWLPLHILNCIT 257

AA2AR_HUMAN AAKSLAIIVGLFALCWLPLHIINCFT 256

AA2BR_HUMAN AAKSLAMIVGIFALCWLPVHAVNCVT 257

AA3R_HUMAN TAKSLFLVLFLFALSWLPLSIINCII 253

TM7

AA1R_HUMAN ILTYIAIFLTHGNSAMNPIVYAFRIQ 293

AA2AR_HUMAN WLMYLAIVLSHTNSVVNPFIYAYRIR 293

AA2BR_HUMAN WAMNMAILLSHANSVVNPIVYAYRNR 295

AA3R_HUMAN LVLYMGILLSHANSMMNPIVYAYKIK 287

Dal Ben et al.

183

Fig. 5. Crystal structure of bovine Rhodopsin (pdb code:

1L9H). Disulfide bond and Retinal are shown. orientation of TM3, TM5, TM6, and TM7 [60].

The rhodopsin crystal structure represented a breakthrough

both in that it demonstrated beyond any doubt the 7 TMH

structure of a G-protein-coupled receptor, assumed several

years earlier, and in providing a much more accurate template

for molecular modeling of adenosine receptors than the

previous projection maps (Fig. 5). Since 2000, other 3D

structures of bovine rhodopsin have been solved through both

X-ray diffraction (pdb 1HZX, 1L9H, and 1U19) and NMR

structure determination (pdb 1JFP, 1LN6), and at the present

the rhodopsin crystal structures are the generally used

templates for building of ARs structures through homology

modeling techniques [61-63].

As first, multiple-sequence alignments of selected G-

protein-coupled receptors are used to collect data about

patterns or motifs present in these sequences, in particular in

the TM regions. The aim is to identify the same patterns into

AR sequences and correctly align them to rhodopsin crystal

structure. A guide for the alignment is the presence of some

key highly conserved amino acid residues, including the DRY

motif (in TM3) and three Pro residues (in TM4, 6 and 7) in the

TM segments of GPCRs. Once aligned the sequences, the

same boundaries are applied for the TM helices of the AR as

are identified from the X-ray crystal structure for the

corresponding sequences of bovine rhodopsin, the Cα

coordinates are used to build the seven TM helices for the

human A3 receptor. Special caution is needed for the second

extracellular (E2) loop, which is described in bovine

rhodopsin to fold back over transmembrane helices, and,

therefore, it limits the size of the active site. Hence, amino

acids of this loop could be involved in direct interactions with

the ligands. The presence of a disulfide bridge between two

cysteine residues in TM3 and E2 may be the driving force to

this peculiar fold of the E2 loop [64].

However, a structure-based approach to adenosine

receptors drug discovery in the absence, but maybe also in the

presence, of the real structures requires a multidisciplinary

approach, where molecular models represent a structural basis

to powerfully integrate experimental data derived from

molecular biological, biophysical, bioinformatics, and

pharmacological methods [13,15]. Indeed, the identification of

discrete ARs regions, or even single amino acids that are

critical for ligand recognition and are responsible for

discriminating between agonist and antagonist ligands, has

been an area of extensive study. In addition to a basic

understanding of receptor activation, it has been hoped that a

delineation of ligand-receptor interaction at a molecular level

may provide the basis for rational drug design. Both TMs and

extracellular regions of ARs have been implicated as playing a

role in the formation of the ligand-binding pocket. Site

directed mutagenesis studies in parallel with different

molecular modeling approaches have been recently used as

powerful strategy to design potent and selective ARs ligands,

and the mutagenesis data are summarized in Table 2. These

data can be very helpful, and indeed recently Jacobson and

collaborators created a “neoceptor” and several constitutively

active mutant human A3 adenosine receptors by site-directed

GPCRs as Therapeutic Targets

184

Table 2. Amino Acids of ARs Implicated in Ligand Recognition-interaction

A1

E16A Agonist affinity reduced 4- to 40-fold with little change in

antagonist affinity [65]

D55A Increase in agonist affinity; no change in antagonist affinity. [65]

P86F Substantial reduction of Ag binding [66]

L88A Substantial reduction of Ag binding [66]

T91A Substantial reduction of Ag binding [66]

Q92A Substantial reduction of Ag binding [66]

S94A Loss of agonist or antagonist binding. Binding restored by

substitution with threonine [65]

C169A/S No detectable radioligand binding [66]

T277A Strong decrease in affinity of NECA. Substitution with serine

notably restored NECA affinity. [67]

A2A

E13Q Decrease in agonist affinity, antagonist binding almost not affected. [68,

69]

V84A/D Loss of agonists and antagonists radioligand binding [66]

T88A Decrease in agonist, but not antagonist, affinity [70]

E151A Loss of agonist and antagonist radioligand binding; strong decrease

in agonist potency. [71]

E169A Loss of agonists and antagonists binding [66]

F182A Loss of agonist and antagonist radioligand binding; replacement

with tryptophan considerably restored binding of ligands [72]

H250A

Loss of agonist and antagonist radioligand binding and agonist

activity; substitution with phenylalanine significantly restored

binding

[72]

N253A Loss of agonist and antagonist radioligand binding [72]

F257A Loss of agonist and antagonist radioligand binding [66]

I274A Considerable decrease in agonist and antagonist radioligand

binding, and in agonist potency [72]

S277A Substantial decrease in agonist affinity; substitution with threonine

restored agonist binding [72]

H278A Substantial loss of agonist and antagonist radioligand binding, and

of agonist potency [72]

H278Y Decrease in agonist affinity, antagonist binding almost not affected. [68]

S281A Loss of agonist and antagonist radioligand binding, and of agonist

activity [72]

A2B

A12T Not affected agonists binding [73]

N273Y Slightly increased affinity for agonists [73]

A3

C88F Strongly reduced activity of agonist [74]

Dal Ben et al.

185

mutagenesis [76,77], which provided new insight into the

molecular recognition in the A3 receptor.

In order to provide additional insights into ligand-A3 AR

interactions, site-directed mutagenesis was used to study the

role of a number of residues in the TM domains and in the

second extracellular loop (EL2) [75]. Several residues of this

receptor within TM domains 3 and 6 and the EL2, which have

been predicted by previous molecular modeling to be involved

in the ligand recognition, were mutated. The N250A mutant

receptor lost the ability to bind both radiolabeled agonist and

antagonist [75]. The H95A mutation considerably reduced the

affinity of both agonists and antagonists. In contrast, the

K152A (EL2), W243A and W243F (TM6) mutations did not

significantly change the agonist binding but decreased

antagonist affinity, suggesting that these residues were critical

for the high affinity of A3 AR antagonists [75]. Although not

important for agonist binding, Trp243 was critical for receptor

activation. The results were successfully interpreted using a

rhodopsin based model of ligand-A3 receptor interactions [78].

Some attempts to set up a high-throughput tools to model

GPCRs have been recently reported. One of these,

“GPCRmod”, firstly proposes a reliable alignment of the

seven transmembrane domains based on pattern/motif

recognition f or each of the 7 TMs that are considered

independently. It then converts the alignment into knowledge-

based three-dimensional (3-D) models starting from a set of 3-

D backbone templates and two separate rotamer libraries for

side chain positioning [79]. Another software, “PREDICT”,

tries to model the 3D structure of GPCRs without relying on

homology to rhodopsin. This algorithm starts from the primary

sequence of the receptor, simultaneously optimizing multiple

conformations of the protein in order to find its most stable

structure, culminating in a virtual receptor-ligand complex

[80].

Combined Target-based and Ligand-based Drug Design Strategy In a recent work, Moro proposes a combined target-based

and ligand-based drug design approach to define a

pharmacophore model of the human A3 receptor antagonists

[81]. This strategy is composed by two different steps; in a

first phase a high throughput molecular docking study is

carried out, and information about the putative binding site

and the binding conformations of some A3 inhibitors are

collected. Secondly, docking based structure superimposition

is used to perform a quantitative study of the structure activity

relationships for binding of these inhibitors to adenosine A3

receptor using a comparative molecular field analysis

(CoMFA). The results are combined to assemble a new target

based pharmacophore model. This method seems to give good

Table 2. Continued.

H95A Considerable decrease in both agonist and antagonist affinity [75]

R108A/K Enhanced potency of agonist [74]

Y109F Strongly reduced activity of agonist [74]

D107K/R108K/E

Complete loss of agonist activity [74]

K152A Agonist binding not affected but rather decreased antagonist affinity [75]

A229E Enhanced potency of agonist [74]

W243A/F Agonist binding not affected but rather decreased antagonist affinity [75]

N250A Loss of agonist and antagonist radioligand binding [75]

H272E Decrease in both agonist and antagonist affinity [76]

Y282F Strongly reduced activity of agonist [74]

GPCRs as Therapeutic Targets

186

results, as the predicted Ki values of new A3 inhibitors were in

agreement with the experimental values.

CONCLUDING REMARKS

G-protein-coupled receptors (GPCRs) compose a big

family of sensory proteins that mediate a wide variety of

physiological processes by responding to different external

stimuli such as hormones, paracrines (local hormones),

neurotransmitters, neuromodulators, odours, and light. These

transmembrane receptors transduce the external signals mostly

(but not only) by interacting with regulatory G-proteins. The

superfamily of GPCRs comprises receptors for many

substances with important physiological functions. For this

reason their dysfunction leads to human diseases, and in fact

many GPCRs are targets for pharmaceuticals and drugs of

abuse. The purine nucleoside adenosine is a mediator playing

an important role in modulating many pathophysiological

conditions in the CNS as well as in peripheral organs and

tissues. Four adenosine receptor subtypes have been identified

by molecular cloning; they belong to the family of GPCRs,

and represent a promising drug target. This is due to the fact

that the receptors are expressed in a large variety of cells, and

there is a large number of ligands, which have been generated

by introducing several modifications in the structure of the

lead compound adenosine some of them highly specific.

Currently, adenosine receptors drug discovery is focused

on the development of more selective and/or potent molecules

that act at the orthosteric site (i.e. the binding site of the

endogenous agonist) of the receptor. Knowledge and mapping

of the structural determinants for optimal receptor function are

essential for understanding the molecular basis of ligand

action and receptor function in normal and abnormal

conditions. Moreover, deciphering structure-function

relationships in adenosine receptors will promote computer-

aided drug discovery by elucidating the binding mode (s) of

known ligands in their receptor binding sites.

REFERENCES

[1] K. Kristiansen, Pharmacol. Ther. 103 (2004) 21.

[2] R.J. Lefkowitz, Trends Pharmacol. Sci. 25 (2004) 413.

[3] J. Bockaert, J.P. Pin, Embo J. 18 (1999) 1723.

[4] T.B. Patel, Pharmacol. Rev. 56 (2004) 371.

[5] J. Bockaert, P. Marin, A. Dumuis, L. Fagni. FEBS Lett.

546 (2003) 65.

[6] T. Schoneberg, G. Schultz, T. Gudermann. Mol. Cell.

Endocrinol. 151 (1999) 181.

[7] K.L. Pierce, R.T. Premont, R.J. Lefkowitz, Nat. Rev.

Mol. Cell. Biol. 3 (2002) 639.

[8] O. El Far, H. Betz, Biochem. J. 365 (2002) 329.

[9] A.L. Lomize, I.D. Pogozheva, H.I. Mosberg, J.

Comput. Aided Mol. Des. 13 (1999) 325.

[10] R. Fredriksson, M.C. Lagerstrom, L.G. Lundin, H.B.

Schioth, Mol. Pharmacol. 63 (2003) 1256.

[11] G. Milligan, Mol. Pharmacol. 66 (2004) 1.

[12] I. Gomes, B.A. Jordan, A. Gupta, C. Rios, N. Trapaidze,

L.A. Devi, J. Mol. Med. 79 (2001) 226.

[13] J. Ballesteros, K. Palczewski, Curr. Opin. Drug Discov.

Devel. 4 (2001) 561.

[14] G. Milligan, G.J. Feng, R.J. Ward, N. Sartania, D.

Ramsay, A.J. McLean, J.J. Carrillo, Curr. Pharm.

Des. 10 (2004) 1989.

[15] O.M. Becker, Y. Marantz, S. Shacham, B. Inbal, A.

Heifetz, O. Kalid, S. Bar-Haim, D. Warshaviak, M.

Fichman, S. Noiman, Proc. Natl. Acad. Sci. USA 101

(2004) 11304.

[16] T. Noji, A. Karasawa, H. Kusaka, Eur. J. Pharmacol.

495 (2004) 1.

[17] a) P.G. Baraldi, B. Cacciari, G. Cristalli, S. Vittori, in:

C. Melchiorre (Ed.), I Recettori Purinergici, Klueb

Editrice, Bologna, 2000; b) M. Klinger, M. Freissmuth,

C. Nanoff, Cell Signal. 14 (2002) 99.

[18] J.A. Ribeiro, A.M. Sebastiao, A. de Mendonca, Prog.

Neurobiol. 68 (2002) 377.

[19] P. Meghji, J.B. Tuttle, R. Rubio, J. Neurochem. 53

(1989) 1852.

[20] B.T. Andresen, D.G. Gillespie, Z. Mi, R.K. Dubey,

E.K. Jackson, J. Pharmacol. Exp. Ther. 291 (1999) 76.

[21] K.K. Kalsi, R.T. Smolenski, M.H. Yacoub, Mol. Cell.

Biochem. 180 (1998) 193.

[22] S. Latini, F. Pedata, J. Neurochem. 79 (2001) 463.

[23] U.K. Decking, G. Schlieper, K. Kroll, J. Schrader, Circ.

Res. 81 (1997) 154.

[24] B.B. Fredholm, M.P. Abbracchio, G. Burnstock, J.W.

Daly, T.K. Harden, K.A. Jacobson, P. Leff, M.

Dal Ben et al.

187

Williams, Pharmacol. Rev. 46 (1994) 143.

[25] I. Feoktistov, I. Biaggioni, Pharmacol. Rev. 49 (1997)

381.

[26] G.F. Baxter, D.M. Yellon, J. Mol. Cell. Cardiol. 31

(1999) 981.

[27] T.C. Zhao, R.C. Kukreja, Am. J. Physiol. Heart Circ.

Physiol. 285 (2003) H434.

[28] K.A. Rudolphi, P. Schubert, F.E. Parkinson, B.B.

Fredholm, Cerebrovasc. Brain Metab. Rev. 4 (1992)

346.

[29] P. Schubert, T. Ogata, C. Marchini, S. Ferroni, K.

Rudolphi, Ann. NY Acad. Sci. 825 (1997) 1.

[30] M.E. Olah, G.L. Stiles, Annu. Rev. Pharmacol. Toxicol.

35 (1995) 581.

[31] a) G. Cristalli, R. Volpini (Eds.), Adenosine Receptors:

Chemistry and Pharmacology, Curr. Top. Med. Chem.

3 (2003); b) G. Cristalli, S. Costanzi, C. Lambertucci,

S. Taffi, S. Vittori, R. Volpini, I1 Farmaco 58 (2003)

193.

[32] C.F. Neely, F.V. Dipierro, M. Kong, J.P. Greelish, T.J.

Gardner, Circulation 94 (1996) II376.

[33] R.F. Bruns, J.J. Katims, Z. Annau, S.H. Snyder,

J.W. Daly, Neuropharmacology 22 (1983) 1523.

[34] J. Sawynok, Eur. J. Pharmacol. 347 (1998) 1.

[35] D. Marx, C.I. Ezeamuzie, K. Nieber, I. Szelenyi, Drug

News Perspect. 14 (2001) 89.

[36] P. Forsythe, L.P. McGarvey, L.G. Heaney, J.

MacMahon, M. Ennis. Clin. Sci. (Lond) 96 (1999) 349.

[37] P.J. Oldenburg, S.J. Mustafa, J. Pharmacol. Exp. Ther.

313 (2005) 319.

[38] a) B.B. Fredholm, Drug News Perspect. 16 (2003) 283;

b) G. Cristalli, C. Lambertucci, S. Taffi, S. Vittori, R.

Volpini, Curr. Top. Med. Chem. 3 (2003) 387; c) R.

Volpini, S. Costanzi, S. Vittori, G. Cristalli, K.N. Klotz,

Curr. Top. Med. Chem. 3 (2003) 427; d) R. Volpini, S.

Costanzi, C. Lambertucci, S. Vittori, K.-N. Klotz, G.

Cristalli, J. Med. Chem. 45 (2002) 3271; e) R. Volpini,

S. Costanzi, C. Lambertucci, S. Vittori, G. Cristalli,

Curr. Pharm. Des. 8 (2002) 2285.

[39] C.F. Neely, I.M. Keith, Am. J. Physiol. 268 (1995)

L1036.

[40] A. Conti, A. Monopoli, M. Gamba, P.A. Borea, E.

Ongini, Naunyn Schmiedebergs Arch. Pharmacol. 348

(1993) 108.

[41] L. Yan, J.C. Burbiel, A. Maass, C.E. Muller, Expert

Opin. Emerg. Drugs 8 (2003) 537.

[42] S. Merighi, P. Mirandola, K. Varani, S. Gessi, E.

Leung, P.G. Baraldi, M.A. Tabrizi, P.A. Borea,

Pharmacol. Ther. 100 (2003) 31.

[43] K.N. Klotz, Naunyn Schmiedebergs Arch. Pharmacol.

362 (2000) 382.

[44] C.E. Muller,T. Scior. Pharm. Acta Helv. 68 (1993) 77.

[45] G. Cristalli, S. Costanzi, C. Lambertucci, G. Lupidi, S.

Vittori, R. Volpini, E. Camaioni. Med. Res. Rev. 21

(2001) 105.

[46] R.M. Berne, Cardiovasc. Res. 27 (1993) 2.

[47] M. Quintana, T. Kahan, P. Hjemdahl, Am. J.

Cardiovasc. Drugs 4 (2004) 159.

[48] A.C. Roach, Crit. Care Nurse 11 (1991) 78.

[49] M.M. LaManna, R. Mohama, I.L. Slavich, 3rd, F.J.

Lumia, S.D. Cha, N. Rambaran, V. Maranhao, Clin.

Nucl. Med. 15 (1990) 804.

[50] P.J. Richardson, A.K. Gubitz, T.C. Freeman, A.K.

Dixon, Adv. Neurol. 80 (1999) 111.

[51] W. Bara-Jimenez, A. Sherzai, T. Dimitrova, A. Favit, F.

Bibbiani, M. Gillespie, M.J. Morris, M.M. Mouradian,

T.N. Chase, Neurology 61 (2003) 293.

[52] A. Pinna, R. Volpini, G. Cristalli, M. Morelli. Eur. J.

Pharmacol. 512 (2005) 157.

[53] R. Henderson, P.N. Unwin, Nature 257 (1975) 28.

[54] G.F. Schertler, C. Villa, R. Henderson, Nature 362

(1993) 770.

[55] V.M. Unger, P.A. Hargrave, J.M. Baldwin, G.F.

Schertler, Nature 389 (1997) 203.

[56] K. Palczewski, T. Kumasaka, T. Hori, C.A. Behnke, H.

Motoshima, B.A. Fox, I. Le Trong, D.C. Teller, T.

Okada, R.E. Stenkamp, M. Yamamoto, M. Miyano,

Science 289 (2000) 739.

[57] P.J. van Galen, A.H. van Bergen, C. Gallo-Rodriguez,

N. Melman, M.E. Olah, I.J. AP, G.L. Stiles, K.A.

Jacobson, Mol. Pharmacol. 45 (1994) 1101.

[58] M.F. Hibert, S. Trumpp-Kallmeyer, A. Bruinvels, J.

Hoflack, Mol. Pharmacol. 40 (1991) 8.

[59] S. Moro, D. Guo, E. Camaioni, J.L. Boyer, T.K. Harden,

K.A. Jacobson, J. Med. Chem. 41 (1998) 1456.

[60] S. Moro, A.H. Li, K.A. Jacobson, J. Chem. Inf.

GPCRs as Therapeutic Targets

188

Comput. Sci. 38 (1998) 1239.

[61] J. Saunders, Drug Discov. Today 6 (2001) 288.

[62] E. Archer, B. Maigret, C. Escrieut, L. Pradayrol, D.

Fourmy, Trends Pharmacol. Sci. 24 (2003) 36.

[63] R.E. Stenkamp, D.C. Teller, K. Palczewski, Arch.

Pharm. (Weinheim) 338 (2005) 209.

[64] a) S. Moro, F. Deflorian, G. Spalluto, G. Pastorin, B.

Cacciari, S.K. Kim, K.A. Jacobson, Chem. Commun.

(Camb) (2003) 2949; b) S. Costanzi, C. Lambertucci, S.

Vittori, R. Volpini, G. Cristalli, J. Mol. Graph. Model.

21 (2003) 253.

[65] H. Barbhaiya, R. McClain, A. Ijzerman, S.A. Rivkees,

Mol. Pharmacol. 50 (1996) 1635.

[66] S.K. Kim, Z.G. Gao, P. Van Rompaey, A.S. Gross,

A. Chen, S. Van Calenbergh, K.A. Jacobson, J. Med.

Chem. 46 (2003) 4847.

[67] A. Townsend-Nicholson, P.R. Schofield, J. Biol. Chem.

269 (1994) 2373.

[68] Z.G. Gao, Q. Jiang, K.A. Jacobson, A.P. Ijzerman,

Biochem. Pharmacol. 60 (2000) 661.

[69] I.J. AP, J.K. Von Frijtag Drabbe Kunzel, J. Kim, Q.

Jiang, K.A. Jacobson, Eur. J. Pharmacol. 310 (1996)

269.

[70] Q. Jiang, A.M. Van Rhee, J. Kim, S. Yehle, J. Wess, K.

A. Jacobson, Mol. Pharmacol. 50 (1996) 512.

[71] J. Kim, Q. Jiang, M. Glashofer, S. Yehle, J. Wess, K.A.

Jacobson, Mol. Pharmacol. 49 (1996) 683.

[72] J. Kim, J. Wess, A.M. van Rhee, T. Schoneberg, K.A.

Jacobson, J. Biol. Chem. 270 (1995) 13987.

[73] M.W. Beukers, H. den Dulk, E.W. van Tilburg, J.

Brouwer, A.P. Ijzerman, Mol. Pharmacol. 58 (2000)

1349.

[74] A. Chen, Z.G. Gao, D. Barak, B.T. Liang, K.A.

Jacobson, Biochem. Biophys. Res. Commun. 284

(2001) 596.

[75] Z.G. Gao, A. Chen, D. Barak, S.K. Kim, C.E. Muller,

K.A. Jacobson, J. Biol. Chem. 277 (2002) 19056.

[76] K.A. Jacobson, Z.G. Gao, A. Chen, D. Barak, S.A.

Kim, K. Lee, A. Link, P.V. Rompaey, S. van

Calenbergh, B.T. Liang, J. Med. Chem. 44 (2001)

4125.

[77] K.A. Jacobson, M. Ohno, H.T. Duong, S.K. Kim,

S. Tchilibon, M. Cesnek, A. Holy, Z.G. Gao, Chem.

Biol. 12 (2005) 237.

[78] Z.G. Gao, S.K. Kim, T. Biadatti, W. Chen, K. Lee,

D. Barak, S.G. Kim, C.R. Johnson, K.A. Jacobson, J.

Med. Chem. 45 (2002) 4471.

[79] C. Bissantz, A. Logean, D. Rognan, J. Chem.

Inf. Comput. Sci. 44 (2004) 1162.

[80] S. Shacham, Y. Marantz, S. Bar-Haim, O. Kalid,

D. Warshaviak, N. Avisar, B. Inbal, A. Heifetz, M.

Fichman, M. Topf, Z. Naor, S. Noiman, O.M. Becker,

Proteins 57 (2004) 51.

[81] S. Moro, P. Braiuca, F. Deflorian, C. Ferrari, G.

Pastorin, B. Cacciari, P.G. Baraldi, K. Varani, P.A.

Borea, G. Spalluto, J. Med. Chem. 48 (2005) 152.