SlideShare a Scribd company logo
1 of 23
Download to read offline
1	
ARE	 P2X	 RECEPTORS	 SUITABLE	 DRUG	 TARGETS	 FOR	 THE	 TREATMENT	 OF	 CHRONIC	
INFLAMMATORY	AND	NEUROPATHIC	PAIN?	
Duuamene	Nyimanu	
MSc	Molecular	Medicine	student,	University	of	East	Anglia,	Norwich	NR4	7TJ	
	
ABSTRACT	
Several	years	ago,	studies	demonstrated	that	extracellular	ATP	is	important	in	
pain	signalling	both	at	the	periphery	and	in	the	CNS.	This	triggered	significant	
advances	in	this	area	resulting	in	the	discovery	of	the	cell-surface	receptor,	P2X	
receptors,	as	ATP-binding	receptors.	It	was	also	found	that	ATP	binding	to	these	
receptors	 results	 in	 their	 activation	 and	 signalling	 in	 different	 pain	 states,	
especially	 chronic	 (inflammatory	 and	 neuropathic)	 pain.	 Inflammatory	 pain	 is	
elicited	 following	 inflammatory	 responses	 to	 peripheral	 nerve	 injury	 or	 an	
unspecific	immune	response,	which	alters	nerve	function.	Generally,	this	type	of	
pain	could	respond	to	treatment	but	neuropathic	pain,	which	develops	following	
nerve	damage	resulting	in	hypersensitivity	in	the	absence	of	overt	stimulus,	is	
usually	 refractory	 to	 treatment.	 Several	 studies	 demonstrated	 that	 ATP-
dependent	 activation	 of	 P2X	 receptors,	 particularly	 P2X3,	 P2X2/3,	 P2X4	 and	
P2X7	receptors,	are	required	for	the	development	of	chronic	inflammatory	and	
neuropathic	 pain,	 and	 that	 blocking	 these	 receptors	 with	 antagonists	 or	
antisense	 oligonucleotide	 silencing	 or	 knockout	 of	 these	 receptors	 in	 mice	
results	in	significant	reduction	in	hypersensitivity	to	pain,	suggesting	that	these	
receptors	 could	 be	 a	 potential	 drug	 target	 in	 managing	 inflammatory	 and	
neuropathic	pain.	This	review	describes	the	latest	evidences	for	the	role	of	P2X	
receptors	 in	 chronic	 inflammatory	 and	 neuropathic	 pain,	 thereby	 establishing	
why	 they	 would	 be	 a	 suitable	 drug	 target	 for	 pain	 management	 and	 conclude	
with	a	review	of	different	drug-like	molecules	that	have	been	tested	in	preclinical	
and	clinical	trial	studies	for	the	treatment	of	these	pain	states.	
	
Introduction	
	
Pain	is	an	unpleasant	sensory	and	emotional	experience	associated	with	actual	
or	 potential	 tissue	 damage.	 It	 minimises	 contact	 with	 the	 injurious	 stimuli	
thereby	promoting	a	protective	response	which	includes	reflex	withdrawal	and	a	
complex	behavioural	strategy	to	avoid	further	pain	[1].	Pain	is	transmitted	via	
the	somatosensory	system,	a	part	of	the	nervous	system,	which	has	evolved	to	
integrate	 sensory	 inputs	 from	 the	 body	 including	 touch,	 heat	 and	 pain	
sensations.	These	sensory	inputs	are	conducted	by	the	primary	afferent	neurons	
on	the	dorsal	side	of	the	spinal	cord,	dorsal	root	ganglion	(DRG)	neurons	from	
the	peripheral	sites	(e.g.	skin)	to	the	dorsal	horn	of	the	spinal	cord,	from	where	
they	are	transmitted	to	the	brain	for	perception	[2],	[3].	Studies	suggest	that	ATP	
released	 by	 activated	 microglia	 in	 sensory	 neurons	 promote	 nociceptor	
signalling	 and	 produces	 fast	 excitatory	 potentials	 in	 the	 dorsal	 root	 ganglion
2	
(DRG)	 neurons	 [4],	 [5].	 Consequently,	 Bleehen	 &	 Keele	 (1977),	 demonstrated	
that	ATP	induced	pain	when	applied	to	a	blister	base	in	human	skin.	This	report	
instigated	enormous	interest	in	the	molecular	mechanism	by	which	ATP	causes	
pain	resulting	in	the	discovery	of	cell-surface	receptors,	thereby	facilitating	the	
detection	of	extracellular	ATP	and	other	nucleotides	on	sensory	neurons	[7].	For	
instance,	it	was	observed	that	ATP	or	its	analogues	in	primary	afferent	neurons	
produce	electrophysiological	and	biological	responses	through	ligand-gated	ion-
channel	 receptors,	 called	 P2X	 receptors	 (P2XRs),	 and	 G	 protein-coupled	
receptors,	 called	 P2Y	 receptors	 (P2YRs)	 [8]–[10].	 However,	 pharmacological	
inhibition	 or	 suppression	 of	 the	 expression	 of	 P2XRs	 or	 P2YRs	 on	 sensory	
neurons	 or	 spinal	 cord	 had	 little	 effect	 on	 acute	 pain	 evoked	 by	 heat	 or	
mechanical	pressure	in	normal	animals	but	inflammatory	pain	was	attenuated	
[11],	 [12],	 suggesting	 that	 the	 actions	 of	 ATP	 and	 its	 receptors	 may	 be	 more	
prominent	in	chronic	pain	especially	inflammatory	and	neuropathic	pain,	than	in	
normal	conditions.	
	
Inflammatory	 pain	 develops	 from	 inflammatory	 responses	 to	 trauma	 in	 the	
peripheral	tissues	and	may	have	physiological	importance	in	that	it	could	assist	
wound	repair	since	contact	with	the	damaged	area	is	minimised	[13]	but	it	could	
also	result	from	non-specific	immune	response	which	alters	nerve	function	[3].	
Also,	inflammatory	pain	may	go	away	after	damage	is	repaired	and	can	generally	
be	 managed	 by	 treatment	 with	 analgesics	 [13].	 However,	 neuropathic	 pain	
usually	 develops	 following	 nerve	 damage,	 which	 may	 be	 caused	 by	 surgery,	
cancer,	bone	compression,	diabetes	or	infection,	but	does	not	resolve	even	when	
the	 damage	 has	 been	 healed	 [1].	 It	 usually	 presents	 as	 hypersensitivity	 in	 the	
absence	 of	 overt	 stimulus	 or	 can	 be	 evoked	 as	 in	 the	 case	 of	 allodynia	 (pain	
resulting	 from	 innocuous	 stimulus)	 and	 hyperalgesia	 (exaggerated	 pain	 in	
response	 to	 noxious	 stimulus),	 and	 is	 often	 refractory	 to	 treatments	 including	
morphines	[1],	[14].	Evidences	suggest	that	the	damage	results	in	the	activation	
of	microglia	cells	in	the	spinal	cord	leading	to	cell	hypertrophy,	proliferation	and	
altered	gene	expression	[7],	[15].	Also,	in	response	to	environmental	factors,	glia	
cells	 evoke	 various	 cellular	 responses	 including	 production	 and	 release	 of	
various	 cytokines	 and	 neurotrophic	 factors	 causing	 neuroanatomical	 and	
neurochemical	transformation	in	the	CNS	that	results	in	the	hyperexcitability	of	
dorsal	 horn	 neurons	 [2],	 [13],	 [16].	 Furthermore,	 several	 studies	 suggest	 that	
P2X3,	P2X4	and	P2X7	receptors	are	important	in	the	pathophysiology	of	chronic	
inflammatory	 and	 neuropathic	 pain	 [15],	 [17]–[19].	 For	 instance,	 it	 had	 been	
reported	 that	 P2X3R	 knockout	 resulted	 in	 enhanced	 thermal	 hyperalgesia	 in	
chronic	inflammation	[11].	Additionally,	it	has	also	been	shown	that	stimulation	
of	P2X4R	results	in	the	release	of	brain-derived	neurotrophic	factor	(BDNF)	and	
a	shift	in	the	neuronal	anion	gradient	in	underlying	neuropathic	pain	[20].
3	
This	review	will	attempt	to	answer	the	question	‘are	P2X	receptors	suitable	drug	
targets	 for	 the	 treatment	 of	 chronic	 inflammatory	 and	 neuropathic	 pain?’	 by	
providing	evidences	for	the	role	of	P2X2/3R	and	P2X3R,	P2X4R,	and	P2X7R	in	
chronic	inflammatory	and	neuropathic	pain.	But	before	this,	it	will	provide	some	
information	 about	 the	 different	 P2X	 receptor	 subtypes	 and	 their	 signalling	
mechanism.	The	review	will	then	discuss	the	success	and	failure	of	experimental	
antagonists	for	these	receptors	and	conclude	with	the	future	perspective	on	P2X	
receptor	targeted	therapies.	
The	P2X	Receptor	subtypes	and	Signalling	
	
The	P2X	family	of	receptors	comprises	seven	subtypes	of	ATP-gated	receptors,	
P2X1-7.	They	were	initially	designated	P2X	by	Burnstock	in	1985	based	on	their	
agonist	and	antagonist	selectivity	in	different	tissues	[21].	This	was	because	ATP	
analogs	 such	 as	 α,β-methylene-ATP	 selectively	 activated	 P2X	 receptors	 while	
adenosine	5’-diphosphate	with	β-sulfur	was	more	selective	for	the	P2Y	receptors	
[22].	It	then	became	clear	that	P2X	receptors,	was	activated	selectively	by	ATP,	
much	 less	 activated	 by	 ADP,	 and	 insensitive	 to	 AMP	 or	 adenosine	 or	 other	
purines	 and	 pyrimidines.	 Additionally,	 this	 family	 of	 receptors	 have	 about	 40-
50%	 amino	 acid	 sequence	 identity	 and	 each	 subunit	 has	 two	 transmembrane	
domains	(TM1	and	TM2),	which	are	separated	by	a	large	extracellular	cysteine-
rich	 domain	 with	 intracellular	 N-terminus	 and	 C-terminus	 of	 considerably	
variable	 length	 [18],	 [22].	 Also,	 the	 channel	 can	 form	 multimers	 of	 several	
subunits	 but	 the	 most	 characterised	 following	 heterologous	 expression	 are	
homomeric	P2X1,	P2X2,	P2X3,	P2X4,	P2X6	and	P2X7	channels,	and	heteromeric	
P2X2/3,	and	P2X1/5.	They	are	abundantly	expressed	in	neurons,	glia,	epithelia,	
endothelial,	 bone,	 muscle	 and	 hematopoietic	 tissues	 and	 they	 are	 involved	 in	
several	 physiological	 processes	 including	 cell	 proliferation,	 differentiation,	
motility	and	death	in	development,	wound	healing,	restenosis	and	epithelial	cell	
turnover	aside	pain	[23],	[24].	
	
Furthermore,	 P2X	 receptors	 mediate	 ATP	 signalling	 mainly	 through	 three	
mechanisms;	 by	 forming	 a	 ligand-gated	 Ca2+-permeable	 cationic	 channels,	
inducing	the	formation	of	a	large	pore,	and	forming	signalling	complexes	with	
interacting	proteins	and	membrane	lipids	[24].	For	instance,	as	a	ligand-gated	
Ca2+-permeable	 cationic	 channel,	 ATP-mediated	 activation	 of	 P2XRs	 has	 been	
found	 to	 induce	 more	 Ca2+	 influx	 than	 glutamate	 ion	 channel	 and	 nicotic	
acetycholine	ion	channels	while	as	large	pore-forming	channel,	some	members	
of	P2X	receptor	family	have	been	shown	to	induce	the	membrane	permeability	
or	pore-formation	upon	prolonged	stimulation	and	these	phenomena	has	been	
observed	in	P2X2,	P2X4,	P2X7,	P2X2/3,	and	P2X2/5	[25];	and	finally	as	signalling	
complexes,	it	has	been	shown	that	P2X	receptors	can	associate	structurally	and	
functionally	with	other	proteins	and	lipids	to	form	ATP	signalling	complexes,	an
4	
example	of	which	is	calmodulin	interaction	with	P2X7	receptor	via	a	calmodulin-
binding	 motif	 to	 form	 a	 signalling	 complex	 necessary	 for	 Ca2+-dependent	
enhancement	of	receptor	activity	and	membrane	blebbing	[24],	[26].	
	
P2X3	And	P2X2/3	Receptors	In	The	Pathogenesis	Of	Inflammatory	And	
Neuropathic	Pain	
	
The	P2X3	receptor	was	the	first	member	of	the	P2X	receptor	family	to	be	cloned	
and	shown	to	be	localised	mainly	on	small	nociceptive	sensory	neurons	in	the	
dorsal	 root	 ganglia	 (DRG)	 [8].	 It	 was	 first	 associated	 with	 pain	 through	 the	
unifying	hypothesis	for	the	initiation	of	pain	[10],	which	stated	that	high	levels	of	
ATP	released	from	tumour	cells	during	abrasive	activity	reaches	P2X3	receptors	
on	 nociceptive	 sensory	 neurons	 in	 the	 DRG	 [27].	 Other	 studies	 later	 used	
immunohistochemical	approach	to	show	that	P2X3	receptors	are	expressed	on	
isolectin	B4	(IB4)	binding	subpopulations	of	small	nociceptive	neurons	and	that	
it	co-localises	with	the	P2X2	receptors	on	large-diameter	 neurons	 in	 the	 DRG,	
forming	a	heteromeric	P2X2/3	receptor	[3].	The	binding	of	ATP	to	the	receptor,	
depolarises	 the	 DRG	 by	 eliciting	 fast-inactivating	 currents	 mediated	 by	 the	
homomeric	P2X3	receptors	while	the	heteromeric	P2X2/3	receptors	were	found	
to	mediate	slow-desensitising	currents	[28].	It	was	also	found	in	DRG	neurons	
isolated	from	rats	with	peripheral	inflammation	induced	by	complete	Freund’s	
adjuvant	(CFA),	that	ATP	application	results	in	the	induction	of	both	fast-	and	
slow-inactivating	currents	in	control	and	inflamed	neurons,	suggesting	that	the	
activation	 of	 this	 receptors	 in	 sensory	 neurons	 facilitates	 the	 transmission	 of	
nociceptive	 signals	 from	 periphery	 to	 the	 spinal	 cord	 [28].	 	 The	 loss	 of	 IB4-
binding	neurons	expressing	P2X3	receptors	resulted	in	decreased	sensitivity	to	
noxious	stimuli	suggesting	a	critical	role	for	these	receptors	in	acute	pain	[27].	
However,	P2X3	and	P2X2/3	receptors	has	now	been	shown	to	play	a	pivotal	role	
in	 the	 signalling	 pathways	 involved	 in	 chronic	 inflammatory	 and	 neuropathic	
pain	[15],	[29].	
	
Several	studies	have	reported	high	levels	P2X3R-mediated	nocifensive	behaviour	
in	 rat	 and	 human	 models	 of	 inflammatory	 pain	 [6],	 [30].	 The	 stimulation	 of	
P2X3R	with	ATP	or	its	analogue	(α,β-methylene-ATP)	in	an	in-vitro-skin-nerve	
model	resulted	in	the	excitation	of	C-mechanoheat	polymodal	nociceptors,	which	
was	 enhanced	 in	 the	 carrageenan-inflamed	 skin	 [31],	 suggesting	 that	 not	 only	
are	 the	 levels	 of	 ATP	 in	 inflamed	 tissues	 elevated	 but	 P2X3	 receptors	 on	 the	
peripheral	nerve	endings	in	inflamed	tissues	could	modulate	pain	transmission.	
Also,	P2X2	knockout	and	P2X2/3	knockout	mice	studies	revealed	that	the	double	
knockout	mice	had	significant	reduction	in	formalin-induced	inflammatory	pain,	
inability	 to	 code	 the	 intensity	 of	 non-noxious	 'warming'	 stimuli,	 inability	 to	
rapidly	desensitise	ATP-induced	currents	in	response	to	ATP	application	as	well
5	
as	decreased	nociceptive	behaviour	compared	to	wildtype	[11],	[32].	Similarly,	
other	 studies	 showed	 that	 P2X3	 antisense	 oligonucleotides	 prevented	
hyperalgesia	 in	 CFA	 model	 of	 chronic	 inflammatory	 pain	 and	 spinal	 nerve	
ligation	model	of	neuropathic	pain,	which	were	correlated	with	decreased	P2X3	
expression	in	the	DRG	[15],	[33].		This	suggests	that	P2X3R	and	P2X2/3R	are	
important	receptors	in	nociceptive	pain	and	that	therapeutically	targeting	them	
with	a	selective	antagonist	could	modulate	pain	state.		
	
Furthermore,	increasing	evidence	suggest	that	persistent	inflammation	by	CFA	is	
accompanied	 by	 upregulation	 of	 both	 P2X2	 and	 P2X3	 receptors	 in	 sensory	
neurons.	 It	 was	 observed	 that	 ATP	 stimulation	 of	 these	 receptors	 in	 inflamed	
DRG	 neurons	 resulted	 in	 elevated	 expression	 of	 P2X2	 and	 P2X3	 receptors	
resulting	 in	 the	 development	 of	 large	 depolarisation	 above	 the	 threshold	 for	
action	 potentials	 compared	 to	 control	 as	 well	 as	 receptor-induced	 increased	
response	 in	 DRG	 neurons	 observed	 in	vitro	 and	 at	 the	 peripheral	 terminals	 in	
vivo	[28].	Also,	in	another	study	it	was	shown	that	intraperitoneal	injection	of	
streptozotocin,	 a	 potent	 P2X3	 agonist,	 in	 a	 diabetic	 neuropathic	 pain	 model	
results	 in	 increased	 membrane	 expression	 of	 P2X3	 receptor	 and	 large	
enhancement	 of	 mechanical	 allodynia,	 which	 was	 significantly	 attenuated	
following	 peripheral	 administration	 of	 P2X3	 receptor	 antagonist,	 pyridoxal-
phosphate-6-azophenyl-2’,4’-disulfonate	 (PPADs)	 and	 TNP-ATP	 [34].	 Similarly,	
using	highly	selective	P2X3	and	P2X2/3	receptor	antagonist	A-317491,	Jarvis	et	
al.	(2002)	showed	that	intraplantar	and	intrathecal	injection	of	A-317491	into	
rats	resulted	in	antinociceptive	effects	in	CFA-induced	chronic	hyperalgesia	and	
nerve	 injury-induced	 hyperalgesia.	 Hence,	 this	 collectively	 demonstrates	 the	
critical	role	of	P2X3	receptors	in	chronic	inflammatory	and	neuropathic	pain	and	
that	 relief	 from	 these	 forms	 of	 pain	 could	 be	 achieved	 by	 pharmacologically	
blocking	P2X3	or	P2X2/3	expression	and/or	activation.		
	
However,	the	cellular	mechanism	by	which	P2X3R	expression	and	function	are	
upregulated	in	sensory	neurons	is	not	fully	known	although	it	is	thought	that	this	
could	 be	 mediated	 by	 interaction	 between	 P2X3	 and	 P2X2/3	 receptors,	 and	
inflammatory	mediators.	This	is	because	various	inflammatory	mediators	such	
as	substance	P,	neurokinin	B,	prostaglandin	E2,	protons	and	bradykinin	strongly	
enhance	 P2X-mediated	 responses	 [7].	 It	 has	 also	 been	 reported	 that	 P2X3	
receptor	activation	in	peripheral	nerve	endings	of	inflamed	tissues	results	in	the	
activation	of	ERK	in	the	DRG	neurons	in	rat	models	of	inflammation	but	not	in	
normal	 rats	 and	 that	 administration	 of	 PPADs	 and	 TNP-ATP,	 significantly	
decreased	the	mechanical	stimulation-evoked	activation	of	ERK	in	CFA-inflamed	
rats	but	not	in	normal	rats	[35].	Moreover,	the	upregulation	of	P2X3	and	P2X2/3	
in	pain	states	have	also	been	associated	with	growth	factors.	For	instance,	it	has	
been	 shown	 that	 glial	 cell	 line-derived	 neurotrophic	 factor	 (GDNF)	 and	 nerve	
growth	factor	(NGF)	treatment	DRG	neurons	increases	the	expression	of	P2X3
6	
receptors,	 with	 evidence	 of	 NGF-mediated	 de	no	P2X3	 expression	 in	 cells	 that	
does	 not	 normally	 express	 the	 receptor,	 suggesting	 a	 mechanism	 of	 NGF-
mediated	 hypersensitivity	 that	 may	 contribute	 to	 chronic	 inflammatory	 pain	
[36].		
P2X4	Receptors	In	The	Pathogenesis	Of	Inflammatory	And	Neuropathic	
Pain	
	
The	 first	 clue	 to	 identifying	 the	 role	 of	 P2X4	 receptors	 in	 the	 spinal	 cord	 in	
neuropathic	 pain	 came	 from	 pharmacological	 investigation	 of	 pain	 behaviour	
after	nerve	injury	using	the	antagonists	TNP-ATP	and	PPADS	[37].	They	reported	
that	 marked	 tactile	 allodynia	 developed	 following	 nerve	 injury	 which	 was	
reversed	 by	 acutely	 administering	 TNP-ATP	 intrathecally	 but	 unaffected	 by	
administering	 PPADS,	 suggesting	 that	 the	 tactile	 allodynia	 depends	 on	 P2X4	
receptors	 in	 the	 spinal	 cord.	 Also,	 immunohistochemical	 analysis	 showed	 that	
many	small	cells,	identified	as	microglia,	in	the	dorsal	horn	of	the	nerve-injured	
side	were	positive	for	P2X4	receptor	protein,	and	showed	high	levels	of	OX-42	
labelling	 and	 morphological	 hypertrophy	 characteristic	 of	 activated	 microglia.	
Additionally,	P2X4	receptor	antisense	oligodeoxynucleotides	(ASO)	reduced	the	
up-regulation	of	P2X4	receptor	protein,	thereby	preventing	the	development	of	
nerve-induced	tractile	allodynia	in	mice	[37].	Moreover,	other	early	studies	in	a	
rat	model	of	neuropathic	pain	induced	by	spinal	nerve	ligation	(SNL)	reported	an	
upregulated	 expression	 of	 P2X4	 receptor	 in	 activated	 spinal	 microglia	 that	
mediate	tactile	allodynia	but	not	in	neurons	[38].	They	observed	that	P2X4KO	
mice	 were	 insensitive	 to	 SNL-induced	 neuropathic	 pain	 experienced	 by	 wild-
type	 littermates.	 This	 collectively	 suggests	 that	 activation	 of	 microglia	 P2X4	
receptor	 is	 necessary	 for	 pain	 hypersensitivity	 following	 nerve	 injury.	
Consequently,	 efforts	 to	 determine	 how	 peripheral	 injury	 increases	 the	
overexpression	 of	 P2X4	 receptor	 in	 microglia	 suggest	 that	 fibronectin	 may	 be	
involved.	It	was	observed	that	microglia	cultured	on	fibronectin-coated	dishes	
showed	 a	 marked	 increase	 in	 P2X4	 receptor	 expression	 at	 both	 mRNA	 and	
protein	 level	 while	 intrathetical	 delivery	 of	 ATP-stimulated	 microglia	 to	 a	 rat	
lumbar	 spinal	 cord,	 showed	 that	 microglia	 treated	 with	 fibronectin	 more	
effectively	 induced	 allodynia	 than	 control	 microglia	 [39].	 Similarly,	 it	 was	
observed	in	a	dorsal	horn	model	of	neuropathic	pain	that	the	level	of	fibronectin	
protein	was	elevated	greatly	after	nerve	injury	as	P2X4	protein	level	increased	
and	pharmacological	inhibition	of	the	fibronectin	receptor	resulted	in	attenuated	
nerve	injury-induced	P2X4	receptor	upregulation	and	pain	hypersensitivity	[40].	
Additionally,	 it	 was	 shown	 in	 Lyn	 tyrosine	 kinase	 knockout	 mice	 studies,	 that	
fibronectin	could	not	induce	the	upregulation	of	P2X4	receptor	in	microglia	cells	
and	neuropathic	pain	in	Lyn-deficient	mice,	suggesting	that	this	kinase	may	be	
important	 in	 the	 molecular	 mechanism	 mediating	 the	 upregulation	 of	 P2X4	
receptors	in	microglia	[41].
7	
	
Furthermore,	Coull	et	al.	(2005)	showed	using	spinal	cord	slices	from	rats	that	
had	 displayed	 pain	 hypersensitivity	 following	 intrathetical	 administration	 of	
P2X4R-stimulated	 microglia,	 	 that	 ATP-stimulated	 microglia	 positively	 shifted	
the	 anion	 reversal	 potential	 (Eanion)	 in	 lamina	 I	 neurons	 and	 rendered	 GABA-
receptor-	 and	 glycine-receptor-mediated	 effects	 depolarising	 rather	 than	
hyperpolarising	 these	 neurons	 (fig.	 1).	 Previously,	 it	 has	 been	 shown	 in	 a	
peripheral	 nerve	 injury	 model	 of	 neuropathic	 pain	 that	 this	 shift	 in	
transmembrane	anion	gradient	which	changes	inhibitory	currents	to	excitatory	
following	nerve	injury,	was	due	to	trans-synaptic	reduction	in	the	expression	of	
the	 potassium-chloride	 exporter	 KCC2	 [42].	 Moreover,	 TNP-ATP	 which	 can	
reverse	nerve-injury	induced	allodynia,	acutely	reverses	the	depolarising	Eanion	
in	the	lamina	I	neurons	after	peripheral	injury	[37].	Therefore,	the	stimulation	of	
P2X4	 receptor	 on	 spinal	 microglia	 causes	 neuropathic	 pain	 through	 increased	
intracellular	chloride	(Cl-)	in	the	spinal	lamina	I	neurons	(fig.1).	
	
	
Fig.	1.	Illustration	of	the	mechanism	by	which	P2X4R	could	modulate	neuropathic	
pain	 [3].	 Damaged	 sensory	 neurons	 release	 ATP,	 which	 binds	 to	 P2X4	 receptor	
resulting	 in	 the	 release	 of	 Ca2+	 and	 activation	 of	 p38	 MAPK,	 which	 induces	 the
8	
release	of	brain	derived	neurotrophic	(BDNF).	The	BDNF	acts	on	its	receptor	Trk	
and	the	inhibitory	interneurons	to	release	GABA.	Also,	action	potentials	from	the	
primary	afferent	terminal	induce	the	release	of	glutamate	and	consequent	opening	
of	AMPA	and	NMDA	receptors.	This	collectively	results	in	the	depolarisation	and	
hyperexcitability	of	the	dorsal	horn	neurons	leading	to	neuropathic	pain.	
Furthermore,	Coull	et	al.	(2005)	also	observed	using	brain	derived	neurotrophic	
factor	 (BDNF)	 administered	 intrathecally	 to	 normal	 rats	 that	 BDNF	 induced	
tactile	allodynia	and	depolarising	shift	in	Eanion	in	lamina	I	neurons	by	peripheral	
nerve	 injury	 comparable	 to	 those	 produced	 by	 ATP-stimulated	 microglia.	
Moreover,	 the	 interruption	 of	 signalling	 between	 BDNF	 and	 its	 receptor	 TrkB,	
either	 by	 pharmacological	 inhibition	 or	 by	 BDNF-sequestering	 fusion	 protein	
(TrkB-Fc)	 prevented	 tactile	 allodynia	 caused	 by	 peripheral	 injury	 or	 by	
intrathecal	 administration	 of	 P2X4-stimulated	 microglia	 [7],	 [20].	 Also,	 it	 was	
observed	that	the	application	of	ATP	to	microglia	induced	the	release	of	BDNF	
but	this	was	abrogated	by	TNP-ATP,	suggesting	that	P2X4R-stimulated	microglia	
release	BDNF	as	a	signalling	factor	leading	to	the	collapse	of	the	transmembrane	
anion	 gradient	 and	 subsequent	 neuronal	 hyperexcitability	 observed	 in	
neuropathic	pain	[3].	Additionally,	in	a	study	involving	P2X4	receptor	knockout	
mice,	 primary	 cultures	 of	 dorsal	 horn	 microglia	 showed	 a	 reduction	 in	 BDNF	
staining	after	ATP	stimulation	in	wild-type	cultures,	while	in	cultures	from	the	
P2X4R-mutant	 mice,	 application	 of	 ATP	 failed	 to	 induce	 any	 change	 [38].	
Similarly,	other	studies	involving	ATP-stimulation	of	P2X4	receptors	resulted	in	
SNARE-mediated	synthesis	and	release	of	BDNF	that	was	dependent	on	the	Ca2+	
influx	through	P2X4	receptors	and	subsequent	p38-MAPK	activation	(fig.	1)	[1],	
[43].	Also,	GABA	receptor-mediated	depolarisation	could	produce	an	excitation	
through	 voltage	 sensitive	 Ca2+	 channels	 and	 NMDA	 receptors	 [3],	 thereby	
suggesting	that	p38-MAPK	as	well	as	GABA	and	NMDA	receptors	are	important	
in	 the	 molecular	 processes	 involved	 in	 P2X4R-mediated	 development	 of	
neuropathic	pain.	
	
Finally,	several	evidences	suggest	that	P2X4	receptors	are	important	in	chronic	
inflammatory	 pain	 development.	 For	 instance,	 P2X4R	 knockout	 mice	 studies	
involving	 the	 injection	 of	 inflammatory	 stimuli	 such	 as	 formalin,	 carrageenan,	
and	CFA	showed	the	complete	loss	of	tactile	allodynia	in	P2X4R-deficient	mice	
compared	 to	 control	 [44],	 [45].	 Also,	 it	 was	 observed	 that	 P2X4R	 deficiency	
attenuates	inflammatory	stimuli-induced	production	of	prostaglandin	E2	(PGE2),	
which	usually	induces	pain	hypersensitivity	by	sensitising	and	overexciting	the	
nociceptive	 neurons,	 from	 macrophages.	 Additionally,	 the	 injection	 of	 naïve	
animals	with	ATP-primed	microglia	or	macrophages	has	been	shown	to	induce	
neuropathic	 and	 chronic	 inflammatory	 pain	 respectively	 [45],	 suggesting	 that	
P2X4	 receptors	 mediate	 the	 cellular	 and	 molecular	 mechanisms	 involved	 in
9	
eliciting	 chronic	 neuropathic	 and	 inflammatory	 pain,	 and	 that	 selectively	
targeting	P2X4	receptors	could	be	a	strategy	for	treatment	of	chronic	pain.	
P2X7	Receptors	In	The	Pathogenesis	Of	Inflammatory	And	Neuropathic	
Pain	
	
P2X7	receptors	are	usually	considered	the	most	unusual	among	the	P2X	receptor	
superfamily	in	terms	of	their	molecular	and	functional	characteristics	due	to	the	
presence	of	additional	200amino	acids	in	their	C-terminal,	and	the	fact	that	aside	
requiring	 high	 ATP	 concentration	 for	 activation,	 prolonged	 agonist	 exposure	
results	 in	 the	 formation	 of	 a	 larger	 pore	 in	 the	 membrane	 [3],	 [16].	 However,	
they	share	a	common	transmembrane	domain	with	other	P2X	receptors.	They	
are	 predominantly	 expressed	 on	 immune	 cells	 including	 lymphocytes	 and	
peripheral	 macrophages	 and	 have	 also	 been	 described	 on	 microglia	 and	
astrocytes.	Like	other	P2XRs,	ATP	binding	activates	the	receptor	resulting	in	the	
opening	of	the	receptor	pore	for	permeation	of	Ca2+,	Na+	and	K+,	which	causes	
changes	in	the	intracellular	concentration	of	potassium	and	consequent	release	
and	activation	of	interleukin-1β	(IL-1β),	a	potent	proinflammatory	cytokine	(fig.	
2)	 [46].	 IL-1β	 induces	 a	 cytokine	 network	 resulting	 in	 the	 production	 of	
superoxide	products,	nitric	oxide	synthase	(iNOS),	cyclo-oxygenase	and	tumour	
necrosis	factor	(TNF)-α,	all	of	which	have	important	roles	in	the	generation	and	
maintenance	of	pain	[17].	Thus,	many	studies	have	been	performed	to	determine	
its	role	in	chronic	inflammatory	and	neuropathic	pain.
10	
	
Fig.	2	The	mechanism	of	action	of	P2X7	receptor	in	IL-1β-mediated	inflammation	
[47].	ATP	binding	to	the	P2X7	receptor	activates	it	resulting	in	the	opening	of	its	
non-selective	ion	pore	and	permeation	of	Ca2+	and	K+.	The	consequent	change	in	
membrane	potential	and	intracellular	Ca2+	and	K+	results	in	the	assembly	of	the	
inflammasome	and	conversion	of	pro-caspase-1	to	active	caspase-1.	Pro-caspase-1	
converts	the	inactive	IL-1β	into	its	active	form	in	the	lysosome,	before	it	is	secreted	
out	of	the	cell.	It	is	also	believed	that	lipopolysaccharide	(LPS)	acts	on	Toll	receptor	
4	 to	 activate	 NFκB	 and	 nuclear	 transcription	 of	 IL-1β,	 later	 translated	 into	 the	
inactive	form	that	is	secreted	into	the	cytoplasm.	
	
P2X7	 receptor	 knockout	 mice	 studies	 have	 facilitated	 the	 investigation	 of	 the	
role	of	this	receptor	in	chronic	pain.	For	instance,	using	P2X7R-/-	mice	models	of	
chronic	 inflammatory	 pain	 (intraplantar	 Freund’s	 complete	 adjuvant)	 and	
neuropathic	pain	(partial	ligation	of	the	sciatic	nerve),	it	was	shown	that	pain	
hypersensitivity	to	both	mechanical	and	thermal	stimuli	was	completely	lost	in	
receptor-deficient	 mice,	 while	 normal	 nociceptive	 processing	 was	 preserved	
[17].	They	also	reported	that	the	receptor	is	upregulated	in	human	dorsal	root	
ganglia	and	injured	nerve	obtained	from	neuropathic	pain	patients,	suggesting	
that	 P2X7	 receptor	 plays	 an	 important	 role	 in	 the	 development	 of	 chronic	
inflammatory	 and	 neuropathic	 pain	 via	 regulation	 of	 IL-1β	 (fig.	 2).	 In	 another	
study	investigating	the	response	of	blood-derived	leukocytes	from	wildtype	and	
P2X7R-/-	mice	to	ATP,	it	was	shown	that	P2X7R-deficiency	results	in	loss	of	ATP-
11	
dependent	 leukocyte	 functions	 including	 IL-1β	 production	 [48].	 They	 also	
showed	 in	 a	 monoclonal	 antibody-induced	 arthritis	 model,	 that	 P2X7R-/-	 was	
associated	 with	 significantly	 attenuated	 arthritis	 compared	 to	 the	 severe	
arthritic	phenotype	observed	in	wildtype	mice,	suggesting	that	ATP-dependent	
activation	of	P2X7	receptor	was	important	in	chronic	inflammatory	pain.	
	
Furthermore,	 the	 pore-forming	 property	 of	 P2X7R	 has	 been	 associated	 with	
development	 of	 chronic	 neuropathic	 pain.	 For	 instance,	 in	 a	 genome-wide	
linkage	 study,	 it	 was	 shown	 that	 mice	 expressing	 P2X7	 receptors	 deficient	 of	
inducing	pore	formation	were	less	sensitive	to	nerve	injury-induced	neuropathic	
pain	than	mice	expressing	the	P2X7	receptors	that	can	induce	pore	formation	
[49].	 The	 study	 also	 found	 that	 within	 two	 cohorts	 of	 patients;	 one	 with	 pain	
after	mastectomy	and	another	cohort	suffering	from	osteoarthritis,	individuals	
expressing	P2X7	receptor	deficient	of	pore	formation	reported	lower	amount	of	
pain	than	those	expressing	the	P2X7	receptor	with	pore-forming	ability.	Also,	the	
administration	of	a	peptide	which	blocks	pore	formation	but	not	channel	activity	
has	been	shown	to	selectively	reduce	nerved	injury	and	inflammatory	allodynia	
in	 wildtype	 mice	 but	 not	 in	 P2X7R-deficient	 mice	 [16].	 This	 suggests	 that	 the	
pore-forming	ability	rather	than	the	small	ion	channel	opening	alone	is	key	to	
the	function	of	P2X7R	in	chronic	inflammatory	and	neuropathic	pain	sensitivity	
and	that	selectively	targeting	pore	formation	could	be	a	strategy	for	treatment	of	
chronic	pain.	
	
Additionally,	aside	its	involvement	in	P2X4R-mediated	allodynia,	p38	MAPK	has	
also	 been	 shown	 to	 mediate	 P2X7R-induced	 production	 of	 IL-1β,	 cathespsin	 S	
and	TNF-α,	which	functions	in	the	maintenance	of	mechanical	hypersensitivity	in	
the	spinal	cord.	Recent	studies	suggest	that	the	phosphorylation	of	p38	MAPK	via	
P2X7	receptor	induce	hyperalgesia	in	an	orofacial	pain	model	following	chronic	
constriction	injury	(CCI)	of	the	infraorbital	nerve	(CCI-ION)	mediated	by	TNF-α	
release	from	microglia	[50].	They	also	observed	that	treatment	of	rats	with	P2X7	
receptor	 agonist,	 3′-O-(4-benzoylbenzoyl)	 adenosine	 5′-triphosphate	 (BzATP),	
induced	tactile	allodynia	through	up-regulation	of	soluble	TNF-α	and	p38	MAPK	
in	 the	 trigeminal	 sensory	 nuclear	 complex	 (TNC)	 which	 was	 inhibited	 by	
SB203580	 (a	 phosphorylated	 p38	 MAPK	 inhibitor)	 and	 Etanercept	 (a	 TNF-α	
inhibitor).	 This	 suggests	 that	 the	 activation	 of	 p38	 MAPK	 could	 be	 a	 possible	
convergence	 point	 in	 the	 P2X4	 and	 P2X7	 receptor	 signalling	 pathways	 during	
neuropathic	pain.	Indeed,	although	not	in	pain	models,	evidence	of	a	structural	
and	functional	interaction	between	the	two	receptors	had	been	described	[51],	
but	it	is	currently	unclear	whether	the	heteromeric	interaction	of	these	receptors	
is	 critical	 in	 chronic	 inflammatory	 or	 neuropathic	 pain.	 Nevertheless,	 the	
expression	of	these	receptors	on	various	cell	types	involved	in	pain	transmission,	
suggests	a	promising	target	for	pharmacological	intervention.	An	example	of	this	
was	 by	 Dell’Antonio	 et	 al.	 (2002)	 who	 demonstrated	 in	 a	 paw	 pressure
12	
experiment	that	an	irreversible	inhibitor	of	P2X7	receptor,	oxidised	ATP,	had	an	
anti-hyperalgesic	effect	on	CFA-induced	mechanical	hyperalgesia.		
	
In	summary,	this	review	have	thus	far	discussed	the	present	evidences	for	the	
important	role	played	by	P2X3,	P2X2/3,	P2X4	and	P2X7	receptors	in	mediating	
ATP	 signalling	 involved	 in	 the	 pathogenesis	 of	 chronic	 inflammatory	 and	
neuropathic	pain.	This	therefore,	suggests	that	these	receptors	are	a	promising	
target	for	pain	therapies.	
P2X	Receptors	as	Therapeutic	Targets	in	Chronic	Inflammatory	and	
Neuropathic	Pain	
	
Several	 attempts	 have	 been	 made	 to	 develop	 molecules	 that	 can	 specifically	
target	 ATP-mediated	 signalling	 through	 different	 members	 of	 P2X	 receptor	
family	involved	in	pain	sensitivity.	Initial	attempts	resulted	in	the	identification	
of	Suramin,	a	large	polysulfonated	molecule,	that	is	active	at	multiple	P2	receptor	
subtypes	 and	 its	 derivatives	 such	 as	 NF023,	 which	 was	 reported	 to	 be	 ~10-
20fold	 more	 selective	 for	 P2X	 receptors;	 NF279	 and	 NF449	 which	 are	 potent	
P2X1	 receptor	 antagonists	 [53].	 Later,	 PPADS,	 a	 potent	 coenzyme	 antagonist	
against	human	P2X1,	P2X7	and	P2Y1	were	discovered	along	with	its	derivatives	
but	the	non-selective	interaction	of	these	compounds	limited	their	progress	as	
potential	 therapeutic	 agents.	 Other	 compounds	 developed	 as	 potential	
antagonists	include	oxidised	ATP,	Brilliant	Blue	G,	KN-62,	NF770	and	NF778	but	
these	compounds	where	unsuccessful	due	to	the	wide	diversity	of	recognition	
sites	and	actions	for	which	ATP	is	a	crucial	ligand	resulting	in	their	non-selective	
interaction	with	the	receptors	[21].	Other	antagonists	developed	to	target	each	
of	these	receptors	are	described	below.	
P2X3	and	P2X2/3	Receptors	
Nucleotide	 derivatives	 were	 developed	 to	 modulate	 the	 activity	 of	 these	
receptors.	Thus,	TNP-ATP,	a	non-selective	but	highly	potent	antagonist	of	P2X1	
and	 P2X3	 receptors	 was	 developed	 and	 shown	 to	 block	 the	 pronociceptive	
effects	 of	 P2X	 receptor	 agonists.	 However,	 the	 ability	 of	 TNP-ATP	 to	 enter	
preclinical	pain	studies	for	management	of	P2X3-mediated	pain	was	limited	by	
its	poor	metabolic	stability	in	the	plasma	[21].	A-317491	is	another	compound	
which	 showed	 high	 capability	 to	 competitively	 block	 homomeric	 P2X3	 and	
heteromeric	 P2X3	 receptors	 when	 administered	 in	 CFA-induced	 inflammatory	
hyperalgesia	 although	 it	 had	 limited	 CNS	 penetration	 following	 systemic	
administration	thereby	requiring	higher	doses	or	intrathecal	administration	to	
effectively	 attenuate	 tactile	 allodynia	 following	 peripheral	 injury	 [12],	 [54].	
Other	 potent	 P2X2/3	 and	 P2X3	 receptor	 antagonists	 have	 been	 identified	
including	RO-4,	reported	to	be	capable	of	crossing	the	blood-brain	barrier	and	
attenuate	 nerve	 injury-induced	 pain	 models,	 and	 has	 significantly	 high	 oral
13	
bioavailability	and	low	plasma	blood	binding	as	well	as	good	CNS	prenetration;	
MK-3901,	 reported	 to	 attenuate	 both	 neuropathic	 and	 chronic	 inflammatory	
pain	 in	 experimental	 models	 [55];	 AZ-2,	 reported	 to	 effectively	 reverse	 CFA-
induced	 mechanical	 allodynia	 following	 systemic	 and	 intraplantar	 dosing	 but	
ineffective	at	intrathecal	dosing;	RO85,	reported	to	have	high	Multi-parameter	
optimization	(MPO)	score	and	oral	bioavailability;	and	finally	AF-219	reported	to	
have	high	antagonists	potency	and	selectivity	for	P2X3	and	P2X2/3	receptors,	
with	 moderate	 protein	 binding	 and	 high	 oral	 bioavailability	 [53].	 However,	
majority	 of	 these	 drug-like	 P2X	 receptor	 antagonists	 were	 unsuccessful	 in	
preclinical	studies	due	to	unsatisfactory	pharmacological	profiles	while	AF-219	
entered	 advanced	 clinical	 trials	 for	 treatment	 of	 osteoarthritic	 knee	 pain	 and	
bladder	pain	[24].	
P2X4	Receptors	
The	 discovery	 of	 potent	 antagonist	 for	 P2X4	 receptors	 is	 still	 in	 its	 infancy	
although	the	crystal	structure	of	the	protein	has	been	solved.	However,	TNP-ATP	
has	been	used	as	putative	antagonists	to	block	P2X4	receptor	activation.	Also,	
Brilliant	 Blue	 G	 is	 also	 believed	 to	 have	 antagonistic	 effects	 on	 the	 receptor	
activity	as	well	as	the	serotonin	reuptake	inhibitor,	paroxetine,	a	clinically	used	
antidepressant	[53],	[56].	Interestingly,	N-(benzyloxycarbonyl)phenoxazine	was	
recently	 discovered	 as	 a	 potent	 and	 selective	 P2X4	 receptor	 antagonist	 that	
could	have	potential	therapeutic	benefit	[57].	
P2X7	Receptors	
The	 increased	 characterisation	 of	 the	 role	 of	 P2X7	 receptor	 in	 different	
inflammatory	 diseases	 not	 limited	 to	 chronic	 pain,	 resulted	 in	 different	
companies	 initiating	 a	 search	 for	 selective	 receptor	 antagonists	 (table	 1).	 This	
search	 resulted	 in	 the	 identification	 of	 AACBA1,	 which	 has	 been	 shown	 to	
attenuate	 collagen-induced	 arthritis	 following	 prophylactic	 dosing	 in	 rats	 and,	
CE-224,535	and	AZD9056	which	failed	phase	IIa	and	phase	IIb	clinical	trial	for	
the	 treatment	 of	 rheumatoid	 arthritic	 pain	 respectively	 for	 lack	 of	 efficacy	
compared	to	control	although	they	had	acceptable	tolerability	and	safety	profile	
[58],	[59].	Also,	systemic	screening	resulted	in	the	discovery	of	other	selective	
antagonists	including	AZ-11645373,	a	highly	potent	P2X7	receptor	antagonists	
which	has	been	shown	to	effectively	inhibit	ATP-	and	Bz-ATP-elicited	currents	as	
well	as	A-438079	and	A-740003,	which	has	been	shown	in	inflammatory	pain	
models	 to	 reduce	 thermal	 hyperalgesia	 with	 some	 success	 [60],	 [61].	 Similar	
effect	was	also	observed	on	experimental	models	of	neuropathic	pain,	an	effect	
that	is	mediated	partly	by	at	least	spinal	and/or	supraspinal	sites	of	action	[62].	
	
																																																								
1	N-(adamantan-1-ylmethyl)-5-[(3R-amino-pyrrolidin-1-yl)methyl]-2-chloro-benzamide, a
hydrochloride salt.
14	
Table	1.	Recently	tested	compounds	with	potential	antagonistic	activity	on	P2X7	
Receptor	[63]	
Compound	 Study	 Trial	
Phase	
Completed	 Observed	
Outcome	
Side	effects	
A-438079	 Neuropathic	
and	
inflammatory	
pain	
Pre-
clinical	
Yes		 Inhibited	
mechanical	
allodynia	 and	
effective	 in	
the	 formalin	
pain	model	
Not	evaluated.	
A-740003	 Inflammatory	
and	
neuropathic	
pain	
Pre-
clinical		
Yes	 Analgesic	
effect	 in	
inflammatory	
and	
neuropathic	
rat	models	
Not	evaluated	
A-804598	 Neuropathic	
and	
inflammatory	
pain	
Pre-
clinical		
Yes		 Analgesic	
effect	 in	
inflammatory	
and	
neuropathic	
rat	models	
Not	evaluated		
A847227	 Inflammatory	
and	
neuropathic	
pain	
Pre-
clinical	
Yes		 Antiallodynic	
effect	 and	
decreased	 IL-
1β	released	in	
mouse	model		
Not	evaluated	
AZ10606120	 Ligand	
interaction	
and	 binding	
to	P2X7R	
Pre-
clinical	
Yes	 Had	 allosteric	
effect	 of	
receptor	
activity	 as	 it	
binds	 non-
ATP	 binding	
site	
Not	evaluated	
AZD9056	 Rheumatoid	
Arthritis	
IIb	 Yes		 No	 significant	
efficacy		
Gastrointestinal	
(vomiting,	
nausea	 and	
diarhoea	
	
GSK314118A	
	
Inflammatory	
pain	
Pre-
clinical	
	
	
Yes		
Analgesic	
effect	 in	 rat	
CFA	 model	 of	
inflammatory	
hyperalgesia	
	
Not	related	
	
Natural	products	as	novel	source	of	analgesics	for	targeting	ATP-mediated	
P2X	receptors	
	
Attempts	to	overcome	the	problem	of	non-selectivity	of	P2X	receptor	antagonists	
resulted	 in	 the	 search	 for	 clues	 from	 natural	 products.	 Natural	 products	 have	
been	 shown	 to	 be	 potentially	 good	 source	 of	 new	 specific	 molecules	 for	 the
15	
treatment	of	different	pain	syndromes.	Thus,	several	natural	products	have	been	
developed	 which	 has	 antagonistic	 properties	 on	 P2X	 receptors	 in	 chronic	
neuropathic	 and	 inflammatory	 pain,	 including	 Emolin,	 Amentoflavone,	
Ligunstrazine,	puerarin	and	purotoxin-1	[64].	For	instance,	it	was	shown	in	rats	
that	 following	 formalin-induced	 pain,	 the	 herbal	 product	 used	 in	 Chinese	
medicine	 called,	 Ligunstrazine	 (tetramethypyrazine)	 derived	 from	 Ligusticum	
wallichii,	 antagonise	 P2X3	 receptor	 resulting	 in	 the	 inhibition	 of	 membrane	
depolarisation	induced	in	DRGs	neurons	[65].	The	same	group	confirmed	this	in	
another	study,	where	they	also	found	that	Ligunstrazine	inhibited	P2X3	receptor	
resulting	in	reduced	ionic	currents	induced	by	ATP	in	the	DRG	neurons	but	it	
was	non-selective	since	it	also	induces	PKC	activation.	Similarly,	in	a	neuropathic	
pain	model	it	was	also	found	that	the	Ligunstrazine	inhibited	the	activation	of	
P2X3	receptors	on	the	primary	afferent	neurons.	
	
Furthermore,	 other	 natural	 products	 including	 puerarin	 were	 discovered	 and	
showed	to	inhibit	burn-associated	hyperalgesia	by	preventing	the	upregulation	
of	P2X3	receptor	expression	in	the	DRG	neurons,	with	similar	analgesic	effect	on	
neuropathic	 pain	 via	 the	 same	 mechanism	 [64].	 Additionally,	 Emodin,	 an	
anthraquinone	 obtained	 from	 rhubarb	 extract	 did	 not	 only	 show	 analgesic	
activity	 on	 neuropathic	 pain	 through	 the	 inhibition	 of	 P2X3	 receptors	 in	 the	
primary	 sensory	 neurons,	 but	 also	 had	 antagonistic	 activity	 on	 the	 P2X7	
receptors	 [64].	 Also,	 a	 peptide	 isolated	 from	 the	 venom	 of	 the	 Asian	 spider	
Geolyscosa	 species,	 Purotoxin,	 has	 been	 shown	 to	 be	 a	 potent	 and	 selective	
antagonist	 of	 P2X3	 receptors,	 as	 it	 selectively	 blocks	 the	 P2X3	 receptor	 ion	
current	 in	 rat	 neurons	 [64].	 Thus,	 these	 evidences	 collectively	 suggest	 that	
natural	 products	 could	 offer	 more	 potent	 and	 selective	 compounds	 for	
therapeutic	targeting	of	P2X	receptors.	
Conclusion	
	
Pain	 is	 the	 unpleasant	 sensory	 and	 emotional	 experience	 associated	 with	 the	
actual	 or	 potential	 tissue	 damage.	 It	 can	 simply	 be	 classified	 into	 acute	 and	
chronic	pain	where	acute	pain	describes	the	actual	pain	sensation	experienced	in	
response	 to	 injury	 or	 tissue	 damage.	 Chronic	 pain,	 which	 can	 further	 be	
subdivided	in	inflammatory	and	neuropathic	pain,	occurs	due	to	neurochemical	
and	 phenotypic	 sensitisation	 of	 the	 peripheral	 central	 sensory	 nerves	
characterised	by	increased	sensitivity	to	painful	stimuli	(hyperalgesia)	and	the	
perception	of	pain	in	response	to	normally	innocuous	stimuli	(allodynia).	Pain	
sensitisation	usually	occurs	in	response	to	tissue	damage	or	inflammation	and	is	
mediated	by	several	pronociceptive	neurotransmitters	and	neurotrophic	factors	
such	as	ATP	or	BDNF.	Extracellular	ATP	release	has	now	been	shown	to	elicit	and	
maintain	 sensations	 following	 inflammation	 and	 nerve	 injury	 by	 activating
16	
homomeric	 and	 heteromeric	 P2X	 receptors	 notably	 P2X2/3,	 P2X3,	 P2X4	 and	
P2X7	 receptors	 on	 the	 peripheral	 nerves	 and	 glial	 cells	 (astrocytes	 and	
microglia)	in	the	spinal	cord.		
	
Furthermore,	several	evidences	suggest	the	invaluable	role	P2X2/3,	P2X3,	P2X4	
and	P2X7	receptors	in	the	molecular	processes	involved	in	pain	sensation,	while	
inhibition	 of	 receptor	 activity	 using	 ASO,	 gene	 knockout	 and	 chemical	
compounds	 collectively	 showed	 that	 the	 absence	 of	 P2X	 receptors	 reverts	 the	
ATP-induced	nociceptive	pain.	Thus,	suggesting	that	inhibition	of	these	receptors	
could	be	a	potential	therapeutic	strategy	for	development	of	future	analgesics.	
Massive	 studies	 in	 this	 area	 have	 resulted	 in	 the	 discovery	 of	 P2XR-targetted	
therapeutics	 or	 molecules.	 These	 P2XR	 antagonists	 include	 MK3901,	 AZ-2,	
A740003,	 AZD9056,	 AZ10606120,	 A847227,	 etc	 which	 have	 been	 shown	 to	
effectively	block	receptor	activity	although	majority	of	them	failed	clinical	trials.	
This	 is	 mostly	 due	 to	 the	 widespread	 expression	 of	 all	 the	 different	 receptor	
subtypes,	which	renders	the	discrimination	between	beneficial	and	side	effects	
extremely	 complicated.	 For	 instance,	 studies	 showed	 that	 double-knockout	
P2X2/P2X3	receptors	in	the	gustatory	nerves	of	mice	eliminates	taste	responses,	
although	the	nerve	was	responsive	to	touch,	temperature	and	menthol	[66],	[67]	
while	P2X4R-/-	in	mice	had	also	been	shown	to	cause	increased	blood	pressure	
and	 decreased	 excretion	 of	 nitric	 oxide	 products	 in	 their	 urine	 compared	 to	
wildtype	 [68].	 Similarly,	 the	 lack	 of	 good	 subtype-selective	 agents,	 and	 when	
selective	ligands	are	available,	the	lack	of	an	acceptable	route	of	administration	
to	humans	are	potential	challenges	to	the	development	of	novel	P2XR	targets.	
Other	 challenges	 include	 the	 restrictive	 tissue	 distribution	 of	 some	 of	 these	
receptors;	 the	 ability	 of	 channels	 to	 form	 heterotrimers,	 the	 ability	 of	 the	
extracellular	 domain	 to	 undergo	 substantial	 conformational	 rearrangement	 on	
channel	 opening	 as	 well	 as	 limited	 information	 on	 the	 role	 of	 P2XRs	 in	
physiological	and	pathological	processes,	although	this	roles	are	now	beginning	
to	be	identified	through	knockout	mice	studies	[21].	However,	the	good	news	is	
that	crystal	structures	of	some	of	these	receptors	are	now	available	in	the	closed	
and	ATP-bound	(open)	states.	Hence,	in	the	nearest	future	we	will	begin	to	fully	
understand	 the	 mechanism	 of	 action	 of	 these	 receptors	 and	 how	 to	
pharmacologically	inhibit	them.	Further	work	could	thus,	explore	the	potential	of	
natural	products	as	possible	source	of	future	P2XR-targeted	therapeutics	as	well	
as	identifying	possible	molecules	that	could	potently	inhibit	P2X4	receptor.	
Reference	
[1]	 T.	Trang,	S.	Beggs,	and	M.	W.	Salter,	“ATP	receptors	gate	microglia	
signaling	in	neuropathic	pain.,”	Exp.	Neurol.,	vol.	234,	no.	2,	pp.	354–61,	
Apr.	2012.
17	
[2]	 V.	Tiwari,	Y.	Guan,	and	S.	N.	Raja,	“Modulating	the	delicate	glial-neuronal	
interactions	in	neuropathic	pain:	promises	and	potential	caveats.,”	
Neurosci.	Biobehav.	Rev.,	vol.	45,	pp.	19–27,	Sep.	2014.	
[3]	 E.	Toulme,	M.	Tsuda,	B.	S.	Khakh,	and	K.	Inoue,	“On	the	Role	of	ATP-Gated	
P2X	Receptors	in	Acute,	Inflammatory	and	Neuropathic	Pain,”	in	
Translational	Pain	Research:	From	Mouse	to	Man,	CRC	Press,	2010.	
[4]	 C.	E.	Jahr	and	T.	M.	Jessell,	“ATP	excites	a	subpopulation	of	rat	dorsal	horn	
neurones,”	Nature,	vol.	304,	no.	5928,	pp.	730–733,	Aug.	1983.	
[5]	 P.	Holton,	“The	liberation	of	adenosine	triphosphate	on	antidromic	
stimulation	of	sensory	nerves.,”	J.	Physiol.,	vol.	145,	no.	3,	pp.	494–504,	
Mar.	1959.	
[6]	 T.	Bleehen	and	C.	A.	Keele,	“Observations	on	the	algogenic	actions	of	
adenosine	compounds	on	the	human	blister	base	preparation.,”	Pain,	vol.	
3,	no.	4,	pp.	367–77,	Aug.	1977.	
[7]	 K.	Inoue,	“P2	receptors	and	chronic	pain.,”	Purinergic	Signal.,	vol.	3,	no.	1–
2,	pp.	135–44,	Mar.	2007.	
[8]	 C.	C.	Chen,	A.	N.	Akopian,	L.	Sivilotti,	D.	Colquhoun,	G.	Burnstock,	and	J.	N.	
Wood,	“A	P2X	purinoceptor	expressed	by	a	subset	of	sensory	neurons.,”	
Nature,	vol.	377,	no.	6548,	pp.	428–31,	Oct.	1995.	
[9]	 V.	Ralevic	and	G.	Burnstock,	“Receptors	for	purines	and	pyrimidines.,”	
Pharmacol.	Rev.,	vol.	50,	no.	3,	pp.	413–92,	Sep.	1998.	
[10]	 G.	Burnstock	and	J.	N.	Wood,	“Purinergic	receptors:	their	role	in	
nociception	and	primary	afferent	neurotransmission,”	Curr.	Opin.	
Neurobiol.,	vol.	6,	no.	4,	pp.	526–532,	Aug.	1996.	
[11]	 V.	Souslova,	P.	Cesare,	Y.	Ding,	A.	N.	Akopian,	L.	Stanfa,	R.	Suzuki,	K.	
Carpenter,	A.	Dickenson,	S.	Boyce,	R.	Hill,	D.	Nebenuis-Oosthuizen,	A.	J.	
Smith,	E.	J.	Kidd,	and	J.	N.	Wood,	“Warm-coding	deficits	and	aberrant	
inflammatory	pain	in	mice	lacking	P2X3	receptors.,”	Nature,	vol.	407,	no.	
6807,	pp.	1015–7,	Oct.	2000.	
[12]	 M.	F.	Jarvis,	E.	C.	Burgard,	S.	McGaraughty,	P.	Honore,	K.	Lynch,	T.	J.	
Brennan,	A.	Subieta,	T.	Van	Biesen,	J.	Cartmell,	B.	Bianchi,	W.	Niforatos,	K.	
Kage,	H.	Yu,	J.	Mikusa,	C.	T.	Wismer,	C.	Z.	Zhu,	K.	Chu,	C.-H.	Lee,	A.	O.	
Stewart,	J.	Polakowski,	B.	F.	Cox,	E.	Kowaluk,	M.	Williams,	J.	Sullivan,	and	C.	
Faltynek,	“A-317491,	a	novel	potent	and	selective	non-nucleotide	
antagonist	of	P2X3	and	P2X2/3	receptors,	reduces	chronic	inflammatory	
and	neuropathic	pain	in	the	rat.,”	Proc.	Natl.	Acad.	Sci.	U.	S.	A.,	vol.	99,	no.	
26,	pp.	17179–84,	Dec.	2002.
18	
[13]	 M.	Tsuda,	H.	Tozaki-Saitoh,	and	K.	Inoue,	“Pain	and	purinergic	signaling.,”	
Brain	Res.	Rev.,	vol.	63,	no.	1–2,	pp.	222–32,	May	2010.	
[14]	 J.	Scholz	and	C.	J.	Woolf,	“Can	we	conquer	pain?,”	Oct.	2002.	
[15]	 P.	Honore,	K.	Kage,	J.	Mikusa,	A.	T.	Watt,	J.	F.	Johnston,	J.	R.	Wyatt,	C.	R.	
Faltynek,	M.	F.	Jarvis,	and	K.	Lynch,	“Analgesic	profile	of	intrathecal	P2X3	
antisense	oligonucleotide	treatment	in	chronic	inflammatory	and	
neuropathic	pain	states	in	rats,”	Pain,	vol.	99,	no.	1–2,	pp.	11–19,	Sep.	
2002.	
[16]	 G.	Magni	and	S.	Ceruti,	“The	Purinergic	System	and	Glial	Cells:	Emerging	
Costars	in	Nociception.,”	Biomed	Res.	Int.,	vol.	2014,	p.	495789,	Jan.	2014.	
[17]	 I.	P.	Chessell,	J.	P.	Hatcher,	C.	Bountra,	A.	D.	Michel,	J.	P.	Hughes,	P.	Green,	J.	
Egerton,	M.	Murfin,	J.	Richardson,	W.	L.	Peck,	C.	B.	A.	Grahames,	M.	A.	
Casula,	Y.	Yiangou,	R.	Birch,	P.	Anand,	and	G.	N.	Buell,	“Disruption	of	the	
P2X7	purinoceptor	gene	abolishes	chronic	inflammatory	and	neuropathic	
pain.,”	Pain,	vol.	114,	no.	3,	pp.	386–96,	Apr.	2005.	
[18]	 A.	Surprenant	and	R.	A.	North,	“Signaling	at	purinergic	P2X	receptors.,”	
Annu.	Rev.	Physiol.,	vol.	71,	pp.	333–59,	Jan.	2009.	
[19]	 S.	McGaraughty,	K.	L.	Chu,	M.	T.	Namovic,	D.	L.	Donnelly-Roberts,	R.	R.	
Harris,	X.-F.	Zhang,	C.-C.	Shieh,	C.	T.	Wismer,	C.	Z.	Zhu,	D.	M.	Gauvin,	A.	C.	
Fabiyi,	P.	Honore,	R.	J.	Gregg,	M.	E.	Kort,	D.	W.	Nelson,	W.	A.	Carroll,	K.	
Marsh,	C.	R.	Faltynek,	and	M.	F.	Jarvis,	“P2X7-related	modulation	of	
pathological	nociception	in	rats.,”	Neuroscience,	vol.	146,	no.	4,	pp.	1817–
28,	Jun.	2007.	
[20]	 J.	A.	M.	Coull,	S.	Beggs,	D.	Boudreau,	D.	Boivin,	M.	Tsuda,	K.	Inoue,	C.	Gravel,	
M.	W.	Salter,	and	Y.	De	Koninck,	“BDNF	from	microglia	causes	the	shift	in	
neuronal	anion	gradient	underlying	neuropathic	pain.,”	Nature,	vol.	438,	
no.	7070,	pp.	1017–21,	Dec.	2005.	
[21]	 R.	A.	North	and	M.	F.	Jarvis,	“P2X	receptors	as	drug	targets.,”	Mol.	
Pharmacol.,	vol.	83,	no.	4,	pp.	759–69,	Apr.	2013.	
[22]	 R.	A.	North,	“Molecular	Physiology	of	P2X	Receptors,”	pp.	1013–1067,	
2002.	
[23]	 M.	P.	Abbracchio,	G.	Burnstock,	A.	Verkhratsky,	and	H.	Zimmermann,	
“Purinergic	signalling	in	the	nervous	system:	an	overview.,”	Trends	
Neurosci.,	vol.	32,	no.	1,	pp.	19–29,	Jan.	2009.	
[24]	 L.-H.	Jiang,	“P2X	receptor-mediated	ATP	purinergic	signaling	in	health	and	
disease,”	Cell	Health	Cytoskelet.,	pp.	83–101,	2012.
19	
[25]	 C.	V,	U.	L,	S.	O,	C.	J,	C.	S,	and	R.	F,	“P2X2	and	P2X5	subunits	define	a	new	
heteromeric	receptor	with	P2X7-like	properties.,”	J	Neurosci,	pp.	4284–96,	
2012.	
[26]	 S.	Roger,	P.	Pelegrin,	and	A.	Surprenant,	“Facilitation	of	P2X7	receptor	
currents	and	membrane	blebbing	via	constitutive	and	dynamic	calmodulin	
binding.,”	J.	Neurosci.,	vol.	28,	no.	25,	pp.	6393–401,	Jun.	2008.	
[27]	 G.	Burnstock,	“Purinergic	P2	receptors	as	targets	for	novel	analgesics.,”	
Pharmacol.	Ther.,	vol.	110,	no.	3,	pp.	433–54,	Jun.	2006.	
[28]	 G.-Y.	Xu	and	L.-Y.	M.	Huang,	“Peripheral	Inflammation	Sensitizes	P2X	
Receptor-Mediated	Responses	in	Rat	Dorsal	Root	Ganglion	Neurons,”	J.	
Neurosci.,	vol.	22,	no.	1,	pp.	93–102,	Jan.	2002.	
[29]	 R.	A.	North,	“P2X3	receptors	and	peripheral	pain	mechanisms.,”	J.	Physiol.,	
vol.	554,	no.	Pt	2,	pp.	301–8,	Jan.	2004.	
[30]	 S.	G.	Hamilton,	J.	Warburton,	A.	Bhattacharjee,	J.	Ward,	and	S.	B.	McMahon,	
“ATP	in	human	skin	elicits	a	dose-related	pain	response	which	is	
potentiated	under	conditions	of	hyperalgesia.,”	Brain,	vol.	123	(	Pt	6,	pp.	
1238–46,	Jun.	2000.	
[31]	 S.	G.	Hamilton,	S.	B.	McMahon,	and	G.	R.	Lewin,	“Selective	activation	of	
nociceptors	by	P2X	receptor	agonists	in	normal	and	inflamed	rat	skin.,”	J.	
Physiol.,	vol.	534,	no.	Pt.	2,	pp.	437–45,	Jul.	2001.	
[32]	 D.	A.	Cockayne,	S.	G.	Hamilton,	Q.	M.	Zhu,	P.	M.	Dunn,	Y.	Zhong,	S.	
Novakovic,	A.	B.	Malmberg,	G.	Cain,	A.	Berson,	L.	Kassotakis,	L.	Hedley,	W.	
G.	Lachnit,	G.	Burnstock,	S.	B.	McMahon,	and	A.	P.	Ford,	“Urinary	bladder	
hyporeflexia	and	reduced	pain-related	behaviour	in	P2X3-deficient	mice.,”	
Nature,	vol.	407,	no.	6807,	pp.	1011–5,	Oct.	2000.	
[33]	 J.	Barclay,	S.	Patel,	G.	Dorn,	G.	Wotherspoon,	S.	Moffatt,	L.	Eunson,	S.	
Abdel’al,	F.	Natt,	J.	Hall,	J.	Winter,	S.	Bevan,	W.	Wishart,	A.	Fox,	and	P.	Ganju,	
“Functional	Downregulation	of	P2X3	Receptor	Subunit	in	Rat	Sensory	
Neurons	Reveals	a	Significant	Role	in	Chronic	Neuropathic	and	
Inflammatory	Pain,”	J.	Neurosci.,	vol.	22,	no.	18,	pp.	8139–8147,	Sep.	2002.	
[34]	 G.-Y.	Xu,	G.	Li,	N.	Liu,	and	L.-Y.	M.	Huang,	“Mechanisms	underlying	
purinergic	P2X3	receptor-mediated	mechanical	allodynia	induced	in	
diabetic	rats.,”	Mol.	Pain,	vol.	7,	p.	60,	Jan.	2011.	
[35]	 Y.	Dai,	T.	Fukuoka,	H.	Wang,	H.	Yamanaka,	K.	Obata,	A.	Tokunaga,	and	K.	
Noguchi,	“Contribution	of	sensitized	P2X	receptors	in	inflamed	tissue	to	
the	mechanical	hypersensitivity	revealed	by	phosphorylated	ERK	in	DRG	
neurons.,”	Pain,	vol.	108,	no.	3,	pp.	258–66,	Apr.	2004.
20	
[36]	 M.	S.	Ramer,	E.	J.	Bradbury,	and	S.	B.	McMahon,	“Nerve	growth	factor	
induces	P2X3	expression	in	sensory	neurons,”	J.	Neurochem.,	vol.	77,	no.	3,	
pp.	864–875,	Dec.	2001.	
[37]	 M.	Tsuda,	Y.	Shigemoto-Mogami,	S.	Koizumi,	A.	Mizokoshi,	S.	Kohsaka,	M.	
W.	Salter,	and	K.	Inoue,	“P2X4	receptors	induced	in	spinal	microglia	gate	
tactile	allodynia	after	nerve	injury.,”	Nature,	vol.	424,	no.	6950,	pp.	778–83,	
Aug.	2003.	
[38]	 L.	Ulmann,	J.	P.	Hatcher,	J.	P.	Hughes,	S.	Chaumont,	P.	J.	Green,	F.	Conquet,	G.	
N.	Buell,	A.	J.	Reeve,	I.	P.	Chessell,	and	F.	Rassendren,	“Up-regulation	of	
P2X4	receptors	in	spinal	microglia	after	peripheral	nerve	injury	mediates	
BDNF	release	and	neuropathic	pain.,”	J.	Neurosci.,	vol.	28,	no.	44,	pp.	
11263–8,	Oct.	2008.	
[39]	 K.	Nasu-Tada,	S.	Koizumi,	M.	Tsuda,	E.	Kunifusa,	and	K.	Inoue,	“Possible	
involvement	of	increase	in	spinal	fibronectin	following	peripheral	nerve	
injury	in	upregulation	of	microglial	P2X4,	a	key	molecule	for	mechanical	
allodynia.,”	Glia,	vol.	53,	no.	7,	pp.	769–75,	May	2006.	
[40]	 M.	Tsuda,	E.	Toyomitsu,	T.	Komatsu,	T.	Masuda,	E.	Kunifusa,	K.	Nasu-Tada,	
S.	Koizumi,	K.	Yamamoto,	J.	Ando,	and	K.	Inoue,	“Fibronectin/integrin	
system	is	involved	in	P2X(4)	receptor	upregulation	in	the	spinal	cord	and	
neuropathic	pain	after	nerve	injury.,”	Glia,	vol.	56,	no.	5,	pp.	579–85,	2008.	
[41]	 M.	Tsuda,	H.	Tozaki-Saitoh,	T.	Masuda,	E.	Toyomitsu,	T.	Tezuka,	T.	
Yamamoto,	and	K.	Inoue,	“Lyn	tyrosine	kinase	is	required	for	P2X(4)	
receptor	upregulation	and	neuropathic	pain	after	peripheral	nerve	
injury.,”	Glia,	vol.	56,	no.	1,	pp.	50–8,	2008.	
[42]	 J.	A.	M.	Coull,	D.	Boudreau,	K.	Bachand,	S.	A.	Prescott,	F.	Nault,	A.	Sík,	P.	De	
Koninck,	and	Y.	De	Koninck,	“Trans-synaptic	shift	in	anion	gradient	in	
spinal	lamina	I	neurons	as	a	mechanism	of	neuropathic	pain.,”	Nature,	vol.	
424,	no.	6951,	pp.	938–42,	Aug.	2003.	
[43]	 T.	Trang,	S.	Beggs,	X.	Wan,	and	M.	W.	Salter,	“P2X4-receptor-mediated	
synthesis	and	release	of	brain-derived	neurotrophic	factor	in	microglia	is	
dependent	on	calcium	and	p38-mitogen-activated	protein	kinase	
activation.,”	J.	Neurosci.,	vol.	29,	no.	11,	pp.	3518–28,	Mar.	2009.	
[44]	 M.	Tsuda,	K.	Kuboyama,	T.	Inoue,	K.	Nagata,	H.	Tozaki-Saitoh,	and	K.	Inoue,	
“Behavioral	phenotypes	of	mice	lacking	purinergic	P2X4	receptors	in	acute	
and	chronic	pain	assays.,”	Mol.	Pain,	vol.	5,	p.	28,	Jan.	2009.	
[45]	 L.	Ulmann,	H.	Hirbec,	and	F.	Rassendren,	“P2X4	receptors	mediate	PGE2	
release	by	tissue-resident	macrophages	and	initiate	inflammatory	pain.,”	
EMBO	J.,	vol.	29,	no.	14,	pp.	2290–300,	Jul.	2010.
21	
[46]	 A.	Colomar,	V.	Marty,	C.	Médina,	C.	Combe,	P.	Parnet,	and	T.	Amédée,	
“Maturation	and	release	of	interleukin-1beta	by	lipopolysaccharide-
primed	mouse	Schwann	cells	require	the	stimulation	of	P2X7	receptors.,”	J.	
Biol.	Chem.,	vol.	278,	no.	33,	pp.	30732–40,	Aug.	2003.	
[47]	 N.	Arulkumaran,	R.	J.	Unwin,	and	F.	W.	Tam,	“A	potential	therapeutic	role	
for	P2X7	receptor	(P2X7R)	antagonists	in	the	treatment	of	inflammatory	
diseases.,”	Expert	Opin.	Investig.	Drugs,	vol.	20,	no.	7,	pp.	897–915,	Jul.	
2011.	
[48]	 J.	M.	Labasi,	N.	Petrushova,	C.	Donovan,	S.	McCurdy,	P.	Lira,	M.	M.	Payette,	
W.	Brissette,	J.	R.	Wicks,	L.	Audoly,	and	C.	A.	Gabel,	“Absence	of	the	P2X7	
receptor	alters	leukocyte	function	and	attenuates	an	inflammatory	
response.,”	J.	Immunol.,	vol.	168,	no.	12,	pp.	6436–45,	Jun.	2002.	
[49]	 R.	E.	Sorge,	T.	Trang,	R.	Dorfman,	S.	B.	Smith,	S.	Beggs,	J.	Ritchie,	J.-S.	Austin,	
D.	V	Zaykin,	H.	Vander	Meulen,	M.	Costigan,	T.	A.	Herbert,	M.	Yarkoni-
Abitbul,	D.	Tichauer,	J.	Livneh,	E.	Gershon,	M.	Zheng,	K.	Tan,	S.	L.	John,	G.	D.	
Slade,	J.	Jordan,	C.	J.	Woolf,	G.	Peltz,	W.	Maixner,	L.	Diatchenko,	Z.	Seltzer,	
M.	W.	Salter,	and	J.	S.	Mogil,	“Genetically	determined	P2X7	receptor	pore	
formation	regulates	variability	in	chronic	pain	sensitivity.,”	Nat.	Med.,	vol.	
18,	no.	4,	pp.	595–9,	Apr.	2012.	
[50]	 G.	Ito,	Y.	Suekawa,	M.	Watanabe,	K.	Takahashi,	T.	Inubushi,	K.	Murasaki,	N.	
Hirose,	S.	Hiyama,	T.	Uchida,	and	K.	Tanne,	“P2X7	receptor	in	the	
trigeminal	sensory	nuclear	complex	contributes	to	tactile	
allodynia/hyperalgesia	following	trigeminal	nerve	injury.,”	Eur.	J.	Pain,	vol.	
17,	no.	2,	pp.	185–99,	Feb.	2013.	
[51]	 C.	Guo,	M.	Masin,	O.	S.	Qureshi,	and	R.	D.	Murrell-Lagnado,	“Evidence	for	
functional	P2X4/P2X7	heteromeric	receptors.,”	Mol.	Pharmacol.,	vol.	72,	
no.	6,	pp.	1447–56,	Dec.	2007.	
[52]	 G.	Dell’Antonio,	A.	Quattrini,	E.	Dal	Cin,	A.	Fulgenzi,	and	M.	E.	Ferrero,	
“Antinociceptive	effect	of	a	new	P2Z/P2X7	antagonist,	oxidized	ATP,	in	
arthritic	rats,”	Neurosci.	Lett.,	vol.	327,	no.	2,	pp.	87–90,	Jul.	2002.	
[53]	 R.	J.	Gum,	B.	Wakefield,	and	M.	F.	Jarvis,	“P2X	receptor	antagonists	for	pain	
management:	examination	of	binding	and	physicochemical	properties.,”	
Purinergic	Signal.,	vol.	8,	no.	Suppl	1,	pp.	41–56,	Feb.	2012.	
[54]	 S.	McGaraughty,	C.	T.	Wismer,	C.	Z.	Zhu,	J.	Mikusa,	P.	Honore,	K.	L.	Chu,	C.-H.	
Lee,	C.	R.	Faltynek,	and	M.	F.	Jarvis,	“Effects	of	A-317491,	a	novel	and	
selective	P2X3/P2X2/3	receptor	antagonist,	on	neuropathic,	inflammatory	
and	chemogenic	nociception	following	intrathecal	and	intraplantar	
administration.,”	Br.	J.	Pharmacol.,	vol.	140,	no.	8,	pp.	1381–8,	Dec.	2003.
22	
[55]	 D.	Donnelly-Roberts,	S.	McGaraughty,	C.-C.	Shieh,	P.	Honore,	and	M.	F.	
Jarvis,	“Painful	purinergic	receptors.,”	J.	Pharmacol.	Exp.	Ther.,	vol.	324,	no.	
2,	pp.	409–15,	Feb.	2008.	
[56]	 E.	Bohn,	S.	Müller,	J.	Lauber,	R.	Geffers,	N.	Speer,	C.	Spieth,	J.	Krejci,	B.	
Manncke,	J.	Buer,	A.	Zell,	and	I.	B.	Autenrieth,	“Gene	expression	patterns	of	
epithelial	cells	modulated	by	pathogenicity	factors	of	Yersinia	
enterocolitica.,”	Cell.	Microbiol.,	vol.	6,	no.	2,	pp.	129–41,	Feb.	2004.	
[57]	 V.	Hernandez-Olmos,	A.	Abdelrahman,	A.	El-Tayeb,	D.	Freudendahl,	S.	
Weinhausen,	and	C.	E.	Müller,	“N-substituted	phenoxazine	and	acridone	
derivatives:	structure-activity	relationships	of	potent	P2X4	receptor	
antagonists.,”	J.	Med.	Chem.,	vol.	55,	no.	22,	pp.	9576–88,	Nov.	2012.	
[58]	 T.	C.	Stock,	B.	J.	Bloom,	N.	Wei,	S.	Ishaq,	W.	Park,	X.	Wang,	P.	Gupta,	and	C.	A.	
Mebus,	“Efficacy	and	safety	of	CE-224,535,	an	antagonist	of	P2X7	receptor,	
in	treatment	of	patients	with	rheumatoid	arthritis	inadequately	controlled	
by	methotrexate.,”	J.	Rheumatol.,	vol.	39,	no.	4,	pp.	720–7,	Apr.	2012.	
[59]	 E.	C.	Keystone,	M.	M.	Wang,	M.	Layton,	S.	Hollis,	and	I.	B.	McInnes,	“Clinical	
evaluation	of	the	efficacy	of	the	P2X7	purinergic	receptor	antagonist	
AZD9056	on	the	signs	and	symptoms	of	rheumatoid	arthritis	in	patients	
with	active	disease	despite	treatment	with	methotrexate	or	
sulphasalazine.,”	Ann.	Rheum.	Dis.,	vol.	71,	no.	10,	pp.	1630–5,	Oct.	2012.	
[60]	 L.	Stokes,	L.-H.	Jiang,	L.	Alcaraz,	J.	Bent,	K.	Bowers,	M.	Fagura,	M.	Furber,	M.	
Mortimore,	M.	Lawson,	J.	Theaker,	C.	Laurent,	M.	Braddock,	and	A.	
Surprenant,	“Characterization	of	a	selective	and	potent	antagonist	of	
human	P2X(7)	receptors,	AZ11645373.,”	Br.	J.	Pharmacol.,	vol.	149,	no.	7,	
pp.	880–7,	Dec.	2006.	
[61]	 P.	Honore,	D.	Donnelly-Roberts,	M.	T.	Namovic,	G.	Hsieh,	C.	Z.	Zhu,	J.	P.	
Mikusa,	G.	Hernandez,	C.	Zhong,	D.	M.	Gauvin,	P.	Chandran,	R.	Harris,	A.	P.	
Medrano,	W.	Carroll,	K.	Marsh,	J.	P.	Sullivan,	C.	R.	Faltynek,	and	M.	F.	Jarvis,	
“A-740003	[N-(1-{[(cyanoimino)(5-quinolinylamino)	methyl]amino}-2,2-
dimethylpropyl)-2-(3,4-dimethoxyphenyl)acetamide],	a	novel	and	
selective	P2X7	receptor	antagonist,	dose-dependently	reduces	
neuropathic	pain	in	the	rat.,”	J.	Pharmacol.	Exp.	Ther.,	vol.	319,	no.	3,	pp.	
1376–85,	Dec.	2006.	
[62]	 K.	Inoue,	“The	function	of	microglia	through	purinergic	receptors:	
neuropathic	pain	and	cytokine	release.,”	Pharmacol.	Ther.,	vol.	109,	no.	1–
2,	pp.	210–26,	Jan.	2006.	
[63]	 L.	A.	Alves,	R.	J.	S.	Bezerra,	R.	X.	Faria,	L.	G.	B.	Ferreira,	and	V.	da	Silva	
Frutuoso,	“Physiological	roles	and	potential	therapeutic	applications	of	the	
P2X7	receptor	in	inflammation	and	pain.,”	Molecules,	vol.	18,	no.	9,	pp.	
10953–72,	Jan.	2013.
23	
[64]	 R.	J.	Soares-Bezerra,	A.	S.	Calheiros,	N.	C.	da	Silva	Ferreira,	V.	da	Silva	
Frutuoso,	and	L.	A.	Alves,	“Natural	Products	as	a	Source	for	New	Anti-
Inflammatory	and	Analgesic	Compounds	through	the	Inhibition	of	
Purinergic	P2X	Receptors.,”	Pharmaceuticals	(Basel).,	vol.	6,	no.	5,	pp.	650–
8,	Jan.	2013.	
[65]	 S.	D.	Liang,	C.	S.	Xu,	T.	Zhou,	H.	Q.	Liu,	Y.	Gao,	and	G.	L.	Li,	
“Tetramethylpyrazine	inhibits	ATP-activated	currents	in	rat	dorsal	root	
ganglion	neurons,”	Brain	Res.,	vol.	1040,	no.	1–2,	pp.	92–97,	2005.	
[66]	 T.	E.	Finger,	V.	Danilova,	J.	Barrows,	D.	L.	Bartel,	A.	J.	Vigers,	L.	Stone,	G.	
Hellekant,	and	S.	C.	Kinnamon,	“ATP	signaling	is	crucial	for	communication	
from	taste	buds	to	gustatory	nerves.,”	Science,	vol.	310,	no.	5753,	pp.	1495–
9,	Dec.	2005.	
[67]	 A.	Vandenbeuch,	C.	B.	Anderson,	J.	Parnes,	K.	Enjyoji,	S.	C.	Robson,	T.	E.	
Finger,	and	S.	C.	Kinnamon,	“Role	of	the	ectonucleotidase	NTPDase2	in	
taste	bud	function.,”	Proc.	Natl.	Acad.	Sci.	U.	S.	A.,	vol.	110,	no.	36,	pp.	
14789–94,	Sep.	2013.	
[68]	 K.	Yamamoto,	T.	Sokabe,	T.	Matsumoto,	K.	Yoshimura,	M.	Shibata,	N.	Ohura,	
T.	Fukuda,	T.	Sato,	K.	Sekine,	S.	Kato,	M.	Isshiki,	T.	Fujita,	M.	Kobayashi,	K.	
Kawamura,	H.	Masuda,	A.	Kamiya,	and	J.	Ando,	“Impaired	flow-dependent	
control	of	vascular	tone	and	remodeling	in	P2X4-deficient	mice.,”	Nat.	
Med.,	vol.	12,	no.	1,	pp.	133–7,	Jan.	2006.

More Related Content

What's hot

Opiod analgesics
Opiod analgesicsOpiod analgesics
Opiod analgesicsKomalantil3
 
Pharmacotherapy of Pain
Pharmacotherapy of PainPharmacotherapy of Pain
Pharmacotherapy of PainDrSnehaDange
 
Current Update in the Management of Post-operative Neuraxial Opioid-induced P...
Current Update in the Management of Post-operative Neuraxial Opioid-induced P...Current Update in the Management of Post-operative Neuraxial Opioid-induced P...
Current Update in the Management of Post-operative Neuraxial Opioid-induced P...asclepiuspdfs
 
Acute postoperative pain
Acute postoperative painAcute postoperative pain
Acute postoperative painSAURABH KAKKAR
 
Post operative pain management
Post operative pain managementPost operative pain management
Post operative pain managementPrasanna Somvanshi
 
inflammatory bio marker for treatment response in first episode psychosis
inflammatory bio marker for treatment response in first episode psychosis inflammatory bio marker for treatment response in first episode psychosis
inflammatory bio marker for treatment response in first episode psychosis Mohamed Ghamry
 
2010 nucynta core speaker slides
2010 nucynta core speaker slides2010 nucynta core speaker slides
2010 nucynta core speaker slidesLoubens Jean-Louis
 
Palmitoylethanolamide in the Treatment of Neuropathic Pain
Palmitoylethanolamide in the Treatment of Neuropathic Pain Palmitoylethanolamide in the Treatment of Neuropathic Pain
Palmitoylethanolamide in the Treatment of Neuropathic Pain Sudhir Kumar
 
Pain definition, pathway,analgesic pathway, types of pain
Pain definition, pathway,analgesic pathway, types of painPain definition, pathway,analgesic pathway, types of pain
Pain definition, pathway,analgesic pathway, types of painekta dwivedi
 
Y2 s1 pain physiology 2014
Y2 s1 pain physiology 2014Y2 s1 pain physiology 2014
Y2 s1 pain physiology 2014vajira54
 
Neuropathic pain lecture by Dr. Rashimul haque
Neuropathic pain lecture  by Dr.  Rashimul haqueNeuropathic pain lecture  by Dr.  Rashimul haque
Neuropathic pain lecture by Dr. Rashimul haqueRashimul haque Rimon
 

What's hot (20)

Opiod analgesics
Opiod analgesicsOpiod analgesics
Opiod analgesics
 
Pharmacotherapy of Pain
Pharmacotherapy of PainPharmacotherapy of Pain
Pharmacotherapy of Pain
 
Current Update in the Management of Post-operative Neuraxial Opioid-induced P...
Current Update in the Management of Post-operative Neuraxial Opioid-induced P...Current Update in the Management of Post-operative Neuraxial Opioid-induced P...
Current Update in the Management of Post-operative Neuraxial Opioid-induced P...
 
Multimodal Regiments for Acute Pain Management - Prof. A. Husni Tanra
Multimodal Regiments for Acute  Pain Management - Prof. A. Husni TanraMultimodal Regiments for Acute  Pain Management - Prof. A. Husni Tanra
Multimodal Regiments for Acute Pain Management - Prof. A. Husni Tanra
 
seminar analgesics
seminar analgesicsseminar analgesics
seminar analgesics
 
Acute postoperative pain
Acute postoperative painAcute postoperative pain
Acute postoperative pain
 
Neuropathic pain
Neuropathic painNeuropathic pain
Neuropathic pain
 
Pain management
Pain management Pain management
Pain management
 
Complex Regional Pain Syndrome
Complex Regional Pain SyndromeComplex Regional Pain Syndrome
Complex Regional Pain Syndrome
 
Post operative pain management
Post operative pain managementPost operative pain management
Post operative pain management
 
Analgesics i
Analgesics  iAnalgesics  i
Analgesics i
 
General anesthetics
General anestheticsGeneral anesthetics
General anesthetics
 
Anti inflammatory in postoperative pain isapm 2015 - dr. S. Gaus
Anti inflammatory in postoperative pain isapm 2015 - dr. S. GausAnti inflammatory in postoperative pain isapm 2015 - dr. S. Gaus
Anti inflammatory in postoperative pain isapm 2015 - dr. S. Gaus
 
inflammatory bio marker for treatment response in first episode psychosis
inflammatory bio marker for treatment response in first episode psychosis inflammatory bio marker for treatment response in first episode psychosis
inflammatory bio marker for treatment response in first episode psychosis
 
2010 nucynta core speaker slides
2010 nucynta core speaker slides2010 nucynta core speaker slides
2010 nucynta core speaker slides
 
Palmitoylethanolamide in the Treatment of Neuropathic Pain
Palmitoylethanolamide in the Treatment of Neuropathic Pain Palmitoylethanolamide in the Treatment of Neuropathic Pain
Palmitoylethanolamide in the Treatment of Neuropathic Pain
 
Pain definition, pathway,analgesic pathway, types of pain
Pain definition, pathway,analgesic pathway, types of painPain definition, pathway,analgesic pathway, types of pain
Pain definition, pathway,analgesic pathway, types of pain
 
Inflamación: nuevas evaluaciones
Inflamación: nuevas evaluacionesInflamación: nuevas evaluaciones
Inflamación: nuevas evaluaciones
 
Y2 s1 pain physiology 2014
Y2 s1 pain physiology 2014Y2 s1 pain physiology 2014
Y2 s1 pain physiology 2014
 
Neuropathic pain lecture by Dr. Rashimul haque
Neuropathic pain lecture  by Dr.  Rashimul haqueNeuropathic pain lecture  by Dr.  Rashimul haque
Neuropathic pain lecture by Dr. Rashimul haque
 

Viewers also liked

Adenosine Receptors
Adenosine ReceptorsAdenosine Receptors
Adenosine ReceptorsStudent
 
Purinergic receptor 1
Purinergic receptor 1Purinergic receptor 1
Purinergic receptor 1Sandhya Talla
 
High throughput screening (HTS) at iNovacia
High throughput screening (HTS) at iNovaciaHigh throughput screening (HTS) at iNovacia
High throughput screening (HTS) at iNovaciaiNovacia AB
 
Adenosine
AdenosineAdenosine
AdenosineJervinM
 
High throughput screening
High throughput screeningHigh throughput screening
High throughput screeningajivengan
 
Glutamate receptor
Glutamate receptorGlutamate receptor
Glutamate receptorAnagha Patil
 
Benzodiazipines
Benzodiazipines  Benzodiazipines
Benzodiazipines havalprit
 
High throughput screening
High throughput screening High throughput screening
High throughput screening RewariBhavya
 
High throughput screening
High throughput screeningHigh throughput screening
High throughput screeningManish Kumar
 

Viewers also liked (12)

Adenosine Receptors
Adenosine ReceptorsAdenosine Receptors
Adenosine Receptors
 
Purinergic receptor 1
Purinergic receptor 1Purinergic receptor 1
Purinergic receptor 1
 
High throughput screening (HTS) at iNovacia
High throughput screening (HTS) at iNovaciaHigh throughput screening (HTS) at iNovacia
High throughput screening (HTS) at iNovacia
 
Adenosine
AdenosineAdenosine
Adenosine
 
Adenosine
AdenosineAdenosine
Adenosine
 
High throughput screening
High throughput screeningHigh throughput screening
High throughput screening
 
Glutamate receptor
Glutamate receptorGlutamate receptor
Glutamate receptor
 
Benzodiazipines
Benzodiazipines  Benzodiazipines
Benzodiazipines
 
High throughput screening
High throughput screening High throughput screening
High throughput screening
 
High throughput screening
High throughput screeningHigh throughput screening
High throughput screening
 
Antiplatelet drugs (antithrombotics)
Antiplatelet drugs (antithrombotics)Antiplatelet drugs (antithrombotics)
Antiplatelet drugs (antithrombotics)
 
Benzodiazepines
BenzodiazepinesBenzodiazepines
Benzodiazepines
 

Similar to Role P2X Receptors as Drug targets in inflammatory and neuropathic pain

The role of the central nervous system in the generation and maintenance of c...
The role of the central nervous system in thegeneration and maintenance of c...The role of the central nervous system in thegeneration and maintenance of c...
The role of the central nervous system in the generation and maintenance of c...Danial Mehranfard
 
d3. Substance P (SP).pdf
d3. Substance P (SP).pdfd3. Substance P (SP).pdf
d3. Substance P (SP).pdfVISHALJADHAV100
 
Anatomy And Physiology Of Pain
Anatomy And Physiology Of PainAnatomy And Physiology Of Pain
Anatomy And Physiology Of PainSteven Wallach
 
Evaluation of analgesic agents
Evaluation of  analgesic agentsEvaluation of  analgesic agents
Evaluation of analgesic agentspurnendu mandal
 
Current therapies in management of neuropathic pain
Current therapies in management of neuropathic painCurrent therapies in management of neuropathic pain
Current therapies in management of neuropathic painpurnendu mandal
 
neuroinflammation
neuroinflammationneuroinflammation
neuroinflammationsarvesh022
 
Chronic pain management
Chronic pain management Chronic pain management
Chronic pain management Kaveri Chozhan
 
Recent advances in Pain treatment
Recent advances in Pain treatmentRecent advances in Pain treatment
Recent advances in Pain treatmentDr. Ashutosh Tiwari
 
Pain Pathophysiology - The Role of Neurotrophic Interactions
Pain Pathophysiology -  The Role of Neurotrophic InteractionsPain Pathophysiology -  The Role of Neurotrophic Interactions
Pain Pathophysiology - The Role of Neurotrophic InteractionsAde Wijaya
 
Fibular (Peroneal) Nerve Entrapment Neuropathy-Review_Crimson Publishers
Fibular (Peroneal) Nerve Entrapment Neuropathy-Review_Crimson PublishersFibular (Peroneal) Nerve Entrapment Neuropathy-Review_Crimson Publishers
Fibular (Peroneal) Nerve Entrapment Neuropathy-Review_Crimson PublishersCrimsonPublishersTNN
 
Hanipsych, pain & dep
Hanipsych, pain & depHanipsych, pain & dep
Hanipsych, pain & depHani Hamed
 
Opiod Analgesics.pdf
Opiod Analgesics.pdfOpiod Analgesics.pdf
Opiod Analgesics.pdfAtiqChohan2
 
Neurobiology of depression- recent updates
Neurobiology of depression- recent updatesNeurobiology of depression- recent updates
Neurobiology of depression- recent updatesSantanu Ghosh
 

Similar to Role P2X Receptors as Drug targets in inflammatory and neuropathic pain (20)

The role of the central nervous system in the generation and maintenance of c...
The role of the central nervous system in thegeneration and maintenance of c...The role of the central nervous system in thegeneration and maintenance of c...
The role of the central nervous system in the generation and maintenance of c...
 
d3. Substance P (SP).pdf
d3. Substance P (SP).pdfd3. Substance P (SP).pdf
d3. Substance P (SP).pdf
 
Seminar pruritus
Seminar  pruritusSeminar  pruritus
Seminar pruritus
 
Acupuntura y alivio del dolor
Acupuntura y alivio del dolorAcupuntura y alivio del dolor
Acupuntura y alivio del dolor
 
Anatomy And Physiology Of Pain
Anatomy And Physiology Of PainAnatomy And Physiology Of Pain
Anatomy And Physiology Of Pain
 
Evaluation of analgesic agents
Evaluation of  analgesic agentsEvaluation of  analgesic agents
Evaluation of analgesic agents
 
dr priyJc ppt
dr priyJc pptdr priyJc ppt
dr priyJc ppt
 
Current therapies in management of neuropathic pain
Current therapies in management of neuropathic painCurrent therapies in management of neuropathic pain
Current therapies in management of neuropathic pain
 
neuroinflammation
neuroinflammationneuroinflammation
neuroinflammation
 
Chronic pain management
Chronic pain management Chronic pain management
Chronic pain management
 
Recent advances in Pain treatment
Recent advances in Pain treatmentRecent advances in Pain treatment
Recent advances in Pain treatment
 
Inflammation and epilepsy
Inflammation and epilepsyInflammation and epilepsy
Inflammation and epilepsy
 
Pain Pathophysiology - The Role of Neurotrophic Interactions
Pain Pathophysiology -  The Role of Neurotrophic InteractionsPain Pathophysiology -  The Role of Neurotrophic Interactions
Pain Pathophysiology - The Role of Neurotrophic Interactions
 
Fibular (Peroneal) Nerve Entrapment Neuropathy-Review_Crimson Publishers
Fibular (Peroneal) Nerve Entrapment Neuropathy-Review_Crimson PublishersFibular (Peroneal) Nerve Entrapment Neuropathy-Review_Crimson Publishers
Fibular (Peroneal) Nerve Entrapment Neuropathy-Review_Crimson Publishers
 
Hanipsych, pain & dep
Hanipsych, pain & depHanipsych, pain & dep
Hanipsych, pain & dep
 
nsaids.pptx
nsaids.pptxnsaids.pptx
nsaids.pptx
 
Opiod Analgesics.pdf
Opiod Analgesics.pdfOpiod Analgesics.pdf
Opiod Analgesics.pdf
 
Neurobiology of depression- recent updates
Neurobiology of depression- recent updatesNeurobiology of depression- recent updates
Neurobiology of depression- recent updates
 
Neuropathic pain
Neuropathic painNeuropathic pain
Neuropathic pain
 
Analgesics in dentistry
Analgesics in dentistryAnalgesics in dentistry
Analgesics in dentistry
 

Role P2X Receptors as Drug targets in inflammatory and neuropathic pain

  • 1. 1 ARE P2X RECEPTORS SUITABLE DRUG TARGETS FOR THE TREATMENT OF CHRONIC INFLAMMATORY AND NEUROPATHIC PAIN? Duuamene Nyimanu MSc Molecular Medicine student, University of East Anglia, Norwich NR4 7TJ ABSTRACT Several years ago, studies demonstrated that extracellular ATP is important in pain signalling both at the periphery and in the CNS. This triggered significant advances in this area resulting in the discovery of the cell-surface receptor, P2X receptors, as ATP-binding receptors. It was also found that ATP binding to these receptors results in their activation and signalling in different pain states, especially chronic (inflammatory and neuropathic) pain. Inflammatory pain is elicited following inflammatory responses to peripheral nerve injury or an unspecific immune response, which alters nerve function. Generally, this type of pain could respond to treatment but neuropathic pain, which develops following nerve damage resulting in hypersensitivity in the absence of overt stimulus, is usually refractory to treatment. Several studies demonstrated that ATP- dependent activation of P2X receptors, particularly P2X3, P2X2/3, P2X4 and P2X7 receptors, are required for the development of chronic inflammatory and neuropathic pain, and that blocking these receptors with antagonists or antisense oligonucleotide silencing or knockout of these receptors in mice results in significant reduction in hypersensitivity to pain, suggesting that these receptors could be a potential drug target in managing inflammatory and neuropathic pain. This review describes the latest evidences for the role of P2X receptors in chronic inflammatory and neuropathic pain, thereby establishing why they would be a suitable drug target for pain management and conclude with a review of different drug-like molecules that have been tested in preclinical and clinical trial studies for the treatment of these pain states. Introduction Pain is an unpleasant sensory and emotional experience associated with actual or potential tissue damage. It minimises contact with the injurious stimuli thereby promoting a protective response which includes reflex withdrawal and a complex behavioural strategy to avoid further pain [1]. Pain is transmitted via the somatosensory system, a part of the nervous system, which has evolved to integrate sensory inputs from the body including touch, heat and pain sensations. These sensory inputs are conducted by the primary afferent neurons on the dorsal side of the spinal cord, dorsal root ganglion (DRG) neurons from the peripheral sites (e.g. skin) to the dorsal horn of the spinal cord, from where they are transmitted to the brain for perception [2], [3]. Studies suggest that ATP released by activated microglia in sensory neurons promote nociceptor signalling and produces fast excitatory potentials in the dorsal root ganglion
  • 2. 2 (DRG) neurons [4], [5]. Consequently, Bleehen & Keele (1977), demonstrated that ATP induced pain when applied to a blister base in human skin. This report instigated enormous interest in the molecular mechanism by which ATP causes pain resulting in the discovery of cell-surface receptors, thereby facilitating the detection of extracellular ATP and other nucleotides on sensory neurons [7]. For instance, it was observed that ATP or its analogues in primary afferent neurons produce electrophysiological and biological responses through ligand-gated ion- channel receptors, called P2X receptors (P2XRs), and G protein-coupled receptors, called P2Y receptors (P2YRs) [8]–[10]. However, pharmacological inhibition or suppression of the expression of P2XRs or P2YRs on sensory neurons or spinal cord had little effect on acute pain evoked by heat or mechanical pressure in normal animals but inflammatory pain was attenuated [11], [12], suggesting that the actions of ATP and its receptors may be more prominent in chronic pain especially inflammatory and neuropathic pain, than in normal conditions. Inflammatory pain develops from inflammatory responses to trauma in the peripheral tissues and may have physiological importance in that it could assist wound repair since contact with the damaged area is minimised [13] but it could also result from non-specific immune response which alters nerve function [3]. Also, inflammatory pain may go away after damage is repaired and can generally be managed by treatment with analgesics [13]. However, neuropathic pain usually develops following nerve damage, which may be caused by surgery, cancer, bone compression, diabetes or infection, but does not resolve even when the damage has been healed [1]. It usually presents as hypersensitivity in the absence of overt stimulus or can be evoked as in the case of allodynia (pain resulting from innocuous stimulus) and hyperalgesia (exaggerated pain in response to noxious stimulus), and is often refractory to treatments including morphines [1], [14]. Evidences suggest that the damage results in the activation of microglia cells in the spinal cord leading to cell hypertrophy, proliferation and altered gene expression [7], [15]. Also, in response to environmental factors, glia cells evoke various cellular responses including production and release of various cytokines and neurotrophic factors causing neuroanatomical and neurochemical transformation in the CNS that results in the hyperexcitability of dorsal horn neurons [2], [13], [16]. Furthermore, several studies suggest that P2X3, P2X4 and P2X7 receptors are important in the pathophysiology of chronic inflammatory and neuropathic pain [15], [17]–[19]. For instance, it had been reported that P2X3R knockout resulted in enhanced thermal hyperalgesia in chronic inflammation [11]. Additionally, it has also been shown that stimulation of P2X4R results in the release of brain-derived neurotrophic factor (BDNF) and a shift in the neuronal anion gradient in underlying neuropathic pain [20].
  • 3. 3 This review will attempt to answer the question ‘are P2X receptors suitable drug targets for the treatment of chronic inflammatory and neuropathic pain?’ by providing evidences for the role of P2X2/3R and P2X3R, P2X4R, and P2X7R in chronic inflammatory and neuropathic pain. But before this, it will provide some information about the different P2X receptor subtypes and their signalling mechanism. The review will then discuss the success and failure of experimental antagonists for these receptors and conclude with the future perspective on P2X receptor targeted therapies. The P2X Receptor subtypes and Signalling The P2X family of receptors comprises seven subtypes of ATP-gated receptors, P2X1-7. They were initially designated P2X by Burnstock in 1985 based on their agonist and antagonist selectivity in different tissues [21]. This was because ATP analogs such as α,β-methylene-ATP selectively activated P2X receptors while adenosine 5’-diphosphate with β-sulfur was more selective for the P2Y receptors [22]. It then became clear that P2X receptors, was activated selectively by ATP, much less activated by ADP, and insensitive to AMP or adenosine or other purines and pyrimidines. Additionally, this family of receptors have about 40- 50% amino acid sequence identity and each subunit has two transmembrane domains (TM1 and TM2), which are separated by a large extracellular cysteine- rich domain with intracellular N-terminus and C-terminus of considerably variable length [18], [22]. Also, the channel can form multimers of several subunits but the most characterised following heterologous expression are homomeric P2X1, P2X2, P2X3, P2X4, P2X6 and P2X7 channels, and heteromeric P2X2/3, and P2X1/5. They are abundantly expressed in neurons, glia, epithelia, endothelial, bone, muscle and hematopoietic tissues and they are involved in several physiological processes including cell proliferation, differentiation, motility and death in development, wound healing, restenosis and epithelial cell turnover aside pain [23], [24]. Furthermore, P2X receptors mediate ATP signalling mainly through three mechanisms; by forming a ligand-gated Ca2+-permeable cationic channels, inducing the formation of a large pore, and forming signalling complexes with interacting proteins and membrane lipids [24]. For instance, as a ligand-gated Ca2+-permeable cationic channel, ATP-mediated activation of P2XRs has been found to induce more Ca2+ influx than glutamate ion channel and nicotic acetycholine ion channels while as large pore-forming channel, some members of P2X receptor family have been shown to induce the membrane permeability or pore-formation upon prolonged stimulation and these phenomena has been observed in P2X2, P2X4, P2X7, P2X2/3, and P2X2/5 [25]; and finally as signalling complexes, it has been shown that P2X receptors can associate structurally and functionally with other proteins and lipids to form ATP signalling complexes, an
  • 4. 4 example of which is calmodulin interaction with P2X7 receptor via a calmodulin- binding motif to form a signalling complex necessary for Ca2+-dependent enhancement of receptor activity and membrane blebbing [24], [26]. P2X3 And P2X2/3 Receptors In The Pathogenesis Of Inflammatory And Neuropathic Pain The P2X3 receptor was the first member of the P2X receptor family to be cloned and shown to be localised mainly on small nociceptive sensory neurons in the dorsal root ganglia (DRG) [8]. It was first associated with pain through the unifying hypothesis for the initiation of pain [10], which stated that high levels of ATP released from tumour cells during abrasive activity reaches P2X3 receptors on nociceptive sensory neurons in the DRG [27]. Other studies later used immunohistochemical approach to show that P2X3 receptors are expressed on isolectin B4 (IB4) binding subpopulations of small nociceptive neurons and that it co-localises with the P2X2 receptors on large-diameter neurons in the DRG, forming a heteromeric P2X2/3 receptor [3]. The binding of ATP to the receptor, depolarises the DRG by eliciting fast-inactivating currents mediated by the homomeric P2X3 receptors while the heteromeric P2X2/3 receptors were found to mediate slow-desensitising currents [28]. It was also found in DRG neurons isolated from rats with peripheral inflammation induced by complete Freund’s adjuvant (CFA), that ATP application results in the induction of both fast- and slow-inactivating currents in control and inflamed neurons, suggesting that the activation of this receptors in sensory neurons facilitates the transmission of nociceptive signals from periphery to the spinal cord [28]. The loss of IB4- binding neurons expressing P2X3 receptors resulted in decreased sensitivity to noxious stimuli suggesting a critical role for these receptors in acute pain [27]. However, P2X3 and P2X2/3 receptors has now been shown to play a pivotal role in the signalling pathways involved in chronic inflammatory and neuropathic pain [15], [29]. Several studies have reported high levels P2X3R-mediated nocifensive behaviour in rat and human models of inflammatory pain [6], [30]. The stimulation of P2X3R with ATP or its analogue (α,β-methylene-ATP) in an in-vitro-skin-nerve model resulted in the excitation of C-mechanoheat polymodal nociceptors, which was enhanced in the carrageenan-inflamed skin [31], suggesting that not only are the levels of ATP in inflamed tissues elevated but P2X3 receptors on the peripheral nerve endings in inflamed tissues could modulate pain transmission. Also, P2X2 knockout and P2X2/3 knockout mice studies revealed that the double knockout mice had significant reduction in formalin-induced inflammatory pain, inability to code the intensity of non-noxious 'warming' stimuli, inability to rapidly desensitise ATP-induced currents in response to ATP application as well
  • 5. 5 as decreased nociceptive behaviour compared to wildtype [11], [32]. Similarly, other studies showed that P2X3 antisense oligonucleotides prevented hyperalgesia in CFA model of chronic inflammatory pain and spinal nerve ligation model of neuropathic pain, which were correlated with decreased P2X3 expression in the DRG [15], [33]. This suggests that P2X3R and P2X2/3R are important receptors in nociceptive pain and that therapeutically targeting them with a selective antagonist could modulate pain state. Furthermore, increasing evidence suggest that persistent inflammation by CFA is accompanied by upregulation of both P2X2 and P2X3 receptors in sensory neurons. It was observed that ATP stimulation of these receptors in inflamed DRG neurons resulted in elevated expression of P2X2 and P2X3 receptors resulting in the development of large depolarisation above the threshold for action potentials compared to control as well as receptor-induced increased response in DRG neurons observed in vitro and at the peripheral terminals in vivo [28]. Also, in another study it was shown that intraperitoneal injection of streptozotocin, a potent P2X3 agonist, in a diabetic neuropathic pain model results in increased membrane expression of P2X3 receptor and large enhancement of mechanical allodynia, which was significantly attenuated following peripheral administration of P2X3 receptor antagonist, pyridoxal- phosphate-6-azophenyl-2’,4’-disulfonate (PPADs) and TNP-ATP [34]. Similarly, using highly selective P2X3 and P2X2/3 receptor antagonist A-317491, Jarvis et al. (2002) showed that intraplantar and intrathecal injection of A-317491 into rats resulted in antinociceptive effects in CFA-induced chronic hyperalgesia and nerve injury-induced hyperalgesia. Hence, this collectively demonstrates the critical role of P2X3 receptors in chronic inflammatory and neuropathic pain and that relief from these forms of pain could be achieved by pharmacologically blocking P2X3 or P2X2/3 expression and/or activation. However, the cellular mechanism by which P2X3R expression and function are upregulated in sensory neurons is not fully known although it is thought that this could be mediated by interaction between P2X3 and P2X2/3 receptors, and inflammatory mediators. This is because various inflammatory mediators such as substance P, neurokinin B, prostaglandin E2, protons and bradykinin strongly enhance P2X-mediated responses [7]. It has also been reported that P2X3 receptor activation in peripheral nerve endings of inflamed tissues results in the activation of ERK in the DRG neurons in rat models of inflammation but not in normal rats and that administration of PPADs and TNP-ATP, significantly decreased the mechanical stimulation-evoked activation of ERK in CFA-inflamed rats but not in normal rats [35]. Moreover, the upregulation of P2X3 and P2X2/3 in pain states have also been associated with growth factors. For instance, it has been shown that glial cell line-derived neurotrophic factor (GDNF) and nerve growth factor (NGF) treatment DRG neurons increases the expression of P2X3
  • 6. 6 receptors, with evidence of NGF-mediated de no P2X3 expression in cells that does not normally express the receptor, suggesting a mechanism of NGF- mediated hypersensitivity that may contribute to chronic inflammatory pain [36]. P2X4 Receptors In The Pathogenesis Of Inflammatory And Neuropathic Pain The first clue to identifying the role of P2X4 receptors in the spinal cord in neuropathic pain came from pharmacological investigation of pain behaviour after nerve injury using the antagonists TNP-ATP and PPADS [37]. They reported that marked tactile allodynia developed following nerve injury which was reversed by acutely administering TNP-ATP intrathecally but unaffected by administering PPADS, suggesting that the tactile allodynia depends on P2X4 receptors in the spinal cord. Also, immunohistochemical analysis showed that many small cells, identified as microglia, in the dorsal horn of the nerve-injured side were positive for P2X4 receptor protein, and showed high levels of OX-42 labelling and morphological hypertrophy characteristic of activated microglia. Additionally, P2X4 receptor antisense oligodeoxynucleotides (ASO) reduced the up-regulation of P2X4 receptor protein, thereby preventing the development of nerve-induced tractile allodynia in mice [37]. Moreover, other early studies in a rat model of neuropathic pain induced by spinal nerve ligation (SNL) reported an upregulated expression of P2X4 receptor in activated spinal microglia that mediate tactile allodynia but not in neurons [38]. They observed that P2X4KO mice were insensitive to SNL-induced neuropathic pain experienced by wild- type littermates. This collectively suggests that activation of microglia P2X4 receptor is necessary for pain hypersensitivity following nerve injury. Consequently, efforts to determine how peripheral injury increases the overexpression of P2X4 receptor in microglia suggest that fibronectin may be involved. It was observed that microglia cultured on fibronectin-coated dishes showed a marked increase in P2X4 receptor expression at both mRNA and protein level while intrathetical delivery of ATP-stimulated microglia to a rat lumbar spinal cord, showed that microglia treated with fibronectin more effectively induced allodynia than control microglia [39]. Similarly, it was observed in a dorsal horn model of neuropathic pain that the level of fibronectin protein was elevated greatly after nerve injury as P2X4 protein level increased and pharmacological inhibition of the fibronectin receptor resulted in attenuated nerve injury-induced P2X4 receptor upregulation and pain hypersensitivity [40]. Additionally, it was shown in Lyn tyrosine kinase knockout mice studies, that fibronectin could not induce the upregulation of P2X4 receptor in microglia cells and neuropathic pain in Lyn-deficient mice, suggesting that this kinase may be important in the molecular mechanism mediating the upregulation of P2X4 receptors in microglia [41].
  • 7. 7 Furthermore, Coull et al. (2005) showed using spinal cord slices from rats that had displayed pain hypersensitivity following intrathetical administration of P2X4R-stimulated microglia, that ATP-stimulated microglia positively shifted the anion reversal potential (Eanion) in lamina I neurons and rendered GABA- receptor- and glycine-receptor-mediated effects depolarising rather than hyperpolarising these neurons (fig. 1). Previously, it has been shown in a peripheral nerve injury model of neuropathic pain that this shift in transmembrane anion gradient which changes inhibitory currents to excitatory following nerve injury, was due to trans-synaptic reduction in the expression of the potassium-chloride exporter KCC2 [42]. Moreover, TNP-ATP which can reverse nerve-injury induced allodynia, acutely reverses the depolarising Eanion in the lamina I neurons after peripheral injury [37]. Therefore, the stimulation of P2X4 receptor on spinal microglia causes neuropathic pain through increased intracellular chloride (Cl-) in the spinal lamina I neurons (fig.1). Fig. 1. Illustration of the mechanism by which P2X4R could modulate neuropathic pain [3]. Damaged sensory neurons release ATP, which binds to P2X4 receptor resulting in the release of Ca2+ and activation of p38 MAPK, which induces the
  • 8. 8 release of brain derived neurotrophic (BDNF). The BDNF acts on its receptor Trk and the inhibitory interneurons to release GABA. Also, action potentials from the primary afferent terminal induce the release of glutamate and consequent opening of AMPA and NMDA receptors. This collectively results in the depolarisation and hyperexcitability of the dorsal horn neurons leading to neuropathic pain. Furthermore, Coull et al. (2005) also observed using brain derived neurotrophic factor (BDNF) administered intrathecally to normal rats that BDNF induced tactile allodynia and depolarising shift in Eanion in lamina I neurons by peripheral nerve injury comparable to those produced by ATP-stimulated microglia. Moreover, the interruption of signalling between BDNF and its receptor TrkB, either by pharmacological inhibition or by BDNF-sequestering fusion protein (TrkB-Fc) prevented tactile allodynia caused by peripheral injury or by intrathecal administration of P2X4-stimulated microglia [7], [20]. Also, it was observed that the application of ATP to microglia induced the release of BDNF but this was abrogated by TNP-ATP, suggesting that P2X4R-stimulated microglia release BDNF as a signalling factor leading to the collapse of the transmembrane anion gradient and subsequent neuronal hyperexcitability observed in neuropathic pain [3]. Additionally, in a study involving P2X4 receptor knockout mice, primary cultures of dorsal horn microglia showed a reduction in BDNF staining after ATP stimulation in wild-type cultures, while in cultures from the P2X4R-mutant mice, application of ATP failed to induce any change [38]. Similarly, other studies involving ATP-stimulation of P2X4 receptors resulted in SNARE-mediated synthesis and release of BDNF that was dependent on the Ca2+ influx through P2X4 receptors and subsequent p38-MAPK activation (fig. 1) [1], [43]. Also, GABA receptor-mediated depolarisation could produce an excitation through voltage sensitive Ca2+ channels and NMDA receptors [3], thereby suggesting that p38-MAPK as well as GABA and NMDA receptors are important in the molecular processes involved in P2X4R-mediated development of neuropathic pain. Finally, several evidences suggest that P2X4 receptors are important in chronic inflammatory pain development. For instance, P2X4R knockout mice studies involving the injection of inflammatory stimuli such as formalin, carrageenan, and CFA showed the complete loss of tactile allodynia in P2X4R-deficient mice compared to control [44], [45]. Also, it was observed that P2X4R deficiency attenuates inflammatory stimuli-induced production of prostaglandin E2 (PGE2), which usually induces pain hypersensitivity by sensitising and overexciting the nociceptive neurons, from macrophages. Additionally, the injection of naïve animals with ATP-primed microglia or macrophages has been shown to induce neuropathic and chronic inflammatory pain respectively [45], suggesting that P2X4 receptors mediate the cellular and molecular mechanisms involved in
  • 9. 9 eliciting chronic neuropathic and inflammatory pain, and that selectively targeting P2X4 receptors could be a strategy for treatment of chronic pain. P2X7 Receptors In The Pathogenesis Of Inflammatory And Neuropathic Pain P2X7 receptors are usually considered the most unusual among the P2X receptor superfamily in terms of their molecular and functional characteristics due to the presence of additional 200amino acids in their C-terminal, and the fact that aside requiring high ATP concentration for activation, prolonged agonist exposure results in the formation of a larger pore in the membrane [3], [16]. However, they share a common transmembrane domain with other P2X receptors. They are predominantly expressed on immune cells including lymphocytes and peripheral macrophages and have also been described on microglia and astrocytes. Like other P2XRs, ATP binding activates the receptor resulting in the opening of the receptor pore for permeation of Ca2+, Na+ and K+, which causes changes in the intracellular concentration of potassium and consequent release and activation of interleukin-1β (IL-1β), a potent proinflammatory cytokine (fig. 2) [46]. IL-1β induces a cytokine network resulting in the production of superoxide products, nitric oxide synthase (iNOS), cyclo-oxygenase and tumour necrosis factor (TNF)-α, all of which have important roles in the generation and maintenance of pain [17]. Thus, many studies have been performed to determine its role in chronic inflammatory and neuropathic pain.
  • 10. 10 Fig. 2 The mechanism of action of P2X7 receptor in IL-1β-mediated inflammation [47]. ATP binding to the P2X7 receptor activates it resulting in the opening of its non-selective ion pore and permeation of Ca2+ and K+. The consequent change in membrane potential and intracellular Ca2+ and K+ results in the assembly of the inflammasome and conversion of pro-caspase-1 to active caspase-1. Pro-caspase-1 converts the inactive IL-1β into its active form in the lysosome, before it is secreted out of the cell. It is also believed that lipopolysaccharide (LPS) acts on Toll receptor 4 to activate NFκB and nuclear transcription of IL-1β, later translated into the inactive form that is secreted into the cytoplasm. P2X7 receptor knockout mice studies have facilitated the investigation of the role of this receptor in chronic pain. For instance, using P2X7R-/- mice models of chronic inflammatory pain (intraplantar Freund’s complete adjuvant) and neuropathic pain (partial ligation of the sciatic nerve), it was shown that pain hypersensitivity to both mechanical and thermal stimuli was completely lost in receptor-deficient mice, while normal nociceptive processing was preserved [17]. They also reported that the receptor is upregulated in human dorsal root ganglia and injured nerve obtained from neuropathic pain patients, suggesting that P2X7 receptor plays an important role in the development of chronic inflammatory and neuropathic pain via regulation of IL-1β (fig. 2). In another study investigating the response of blood-derived leukocytes from wildtype and P2X7R-/- mice to ATP, it was shown that P2X7R-deficiency results in loss of ATP-
  • 11. 11 dependent leukocyte functions including IL-1β production [48]. They also showed in a monoclonal antibody-induced arthritis model, that P2X7R-/- was associated with significantly attenuated arthritis compared to the severe arthritic phenotype observed in wildtype mice, suggesting that ATP-dependent activation of P2X7 receptor was important in chronic inflammatory pain. Furthermore, the pore-forming property of P2X7R has been associated with development of chronic neuropathic pain. For instance, in a genome-wide linkage study, it was shown that mice expressing P2X7 receptors deficient of inducing pore formation were less sensitive to nerve injury-induced neuropathic pain than mice expressing the P2X7 receptors that can induce pore formation [49]. The study also found that within two cohorts of patients; one with pain after mastectomy and another cohort suffering from osteoarthritis, individuals expressing P2X7 receptor deficient of pore formation reported lower amount of pain than those expressing the P2X7 receptor with pore-forming ability. Also, the administration of a peptide which blocks pore formation but not channel activity has been shown to selectively reduce nerved injury and inflammatory allodynia in wildtype mice but not in P2X7R-deficient mice [16]. This suggests that the pore-forming ability rather than the small ion channel opening alone is key to the function of P2X7R in chronic inflammatory and neuropathic pain sensitivity and that selectively targeting pore formation could be a strategy for treatment of chronic pain. Additionally, aside its involvement in P2X4R-mediated allodynia, p38 MAPK has also been shown to mediate P2X7R-induced production of IL-1β, cathespsin S and TNF-α, which functions in the maintenance of mechanical hypersensitivity in the spinal cord. Recent studies suggest that the phosphorylation of p38 MAPK via P2X7 receptor induce hyperalgesia in an orofacial pain model following chronic constriction injury (CCI) of the infraorbital nerve (CCI-ION) mediated by TNF-α release from microglia [50]. They also observed that treatment of rats with P2X7 receptor agonist, 3′-O-(4-benzoylbenzoyl) adenosine 5′-triphosphate (BzATP), induced tactile allodynia through up-regulation of soluble TNF-α and p38 MAPK in the trigeminal sensory nuclear complex (TNC) which was inhibited by SB203580 (a phosphorylated p38 MAPK inhibitor) and Etanercept (a TNF-α inhibitor). This suggests that the activation of p38 MAPK could be a possible convergence point in the P2X4 and P2X7 receptor signalling pathways during neuropathic pain. Indeed, although not in pain models, evidence of a structural and functional interaction between the two receptors had been described [51], but it is currently unclear whether the heteromeric interaction of these receptors is critical in chronic inflammatory or neuropathic pain. Nevertheless, the expression of these receptors on various cell types involved in pain transmission, suggests a promising target for pharmacological intervention. An example of this was by Dell’Antonio et al. (2002) who demonstrated in a paw pressure
  • 12. 12 experiment that an irreversible inhibitor of P2X7 receptor, oxidised ATP, had an anti-hyperalgesic effect on CFA-induced mechanical hyperalgesia. In summary, this review have thus far discussed the present evidences for the important role played by P2X3, P2X2/3, P2X4 and P2X7 receptors in mediating ATP signalling involved in the pathogenesis of chronic inflammatory and neuropathic pain. This therefore, suggests that these receptors are a promising target for pain therapies. P2X Receptors as Therapeutic Targets in Chronic Inflammatory and Neuropathic Pain Several attempts have been made to develop molecules that can specifically target ATP-mediated signalling through different members of P2X receptor family involved in pain sensitivity. Initial attempts resulted in the identification of Suramin, a large polysulfonated molecule, that is active at multiple P2 receptor subtypes and its derivatives such as NF023, which was reported to be ~10- 20fold more selective for P2X receptors; NF279 and NF449 which are potent P2X1 receptor antagonists [53]. Later, PPADS, a potent coenzyme antagonist against human P2X1, P2X7 and P2Y1 were discovered along with its derivatives but the non-selective interaction of these compounds limited their progress as potential therapeutic agents. Other compounds developed as potential antagonists include oxidised ATP, Brilliant Blue G, KN-62, NF770 and NF778 but these compounds where unsuccessful due to the wide diversity of recognition sites and actions for which ATP is a crucial ligand resulting in their non-selective interaction with the receptors [21]. Other antagonists developed to target each of these receptors are described below. P2X3 and P2X2/3 Receptors Nucleotide derivatives were developed to modulate the activity of these receptors. Thus, TNP-ATP, a non-selective but highly potent antagonist of P2X1 and P2X3 receptors was developed and shown to block the pronociceptive effects of P2X receptor agonists. However, the ability of TNP-ATP to enter preclinical pain studies for management of P2X3-mediated pain was limited by its poor metabolic stability in the plasma [21]. A-317491 is another compound which showed high capability to competitively block homomeric P2X3 and heteromeric P2X3 receptors when administered in CFA-induced inflammatory hyperalgesia although it had limited CNS penetration following systemic administration thereby requiring higher doses or intrathecal administration to effectively attenuate tactile allodynia following peripheral injury [12], [54]. Other potent P2X2/3 and P2X3 receptor antagonists have been identified including RO-4, reported to be capable of crossing the blood-brain barrier and attenuate nerve injury-induced pain models, and has significantly high oral
  • 13. 13 bioavailability and low plasma blood binding as well as good CNS prenetration; MK-3901, reported to attenuate both neuropathic and chronic inflammatory pain in experimental models [55]; AZ-2, reported to effectively reverse CFA- induced mechanical allodynia following systemic and intraplantar dosing but ineffective at intrathecal dosing; RO85, reported to have high Multi-parameter optimization (MPO) score and oral bioavailability; and finally AF-219 reported to have high antagonists potency and selectivity for P2X3 and P2X2/3 receptors, with moderate protein binding and high oral bioavailability [53]. However, majority of these drug-like P2X receptor antagonists were unsuccessful in preclinical studies due to unsatisfactory pharmacological profiles while AF-219 entered advanced clinical trials for treatment of osteoarthritic knee pain and bladder pain [24]. P2X4 Receptors The discovery of potent antagonist for P2X4 receptors is still in its infancy although the crystal structure of the protein has been solved. However, TNP-ATP has been used as putative antagonists to block P2X4 receptor activation. Also, Brilliant Blue G is also believed to have antagonistic effects on the receptor activity as well as the serotonin reuptake inhibitor, paroxetine, a clinically used antidepressant [53], [56]. Interestingly, N-(benzyloxycarbonyl)phenoxazine was recently discovered as a potent and selective P2X4 receptor antagonist that could have potential therapeutic benefit [57]. P2X7 Receptors The increased characterisation of the role of P2X7 receptor in different inflammatory diseases not limited to chronic pain, resulted in different companies initiating a search for selective receptor antagonists (table 1). This search resulted in the identification of AACBA1, which has been shown to attenuate collagen-induced arthritis following prophylactic dosing in rats and, CE-224,535 and AZD9056 which failed phase IIa and phase IIb clinical trial for the treatment of rheumatoid arthritic pain respectively for lack of efficacy compared to control although they had acceptable tolerability and safety profile [58], [59]. Also, systemic screening resulted in the discovery of other selective antagonists including AZ-11645373, a highly potent P2X7 receptor antagonists which has been shown to effectively inhibit ATP- and Bz-ATP-elicited currents as well as A-438079 and A-740003, which has been shown in inflammatory pain models to reduce thermal hyperalgesia with some success [60], [61]. Similar effect was also observed on experimental models of neuropathic pain, an effect that is mediated partly by at least spinal and/or supraspinal sites of action [62]. 1 N-(adamantan-1-ylmethyl)-5-[(3R-amino-pyrrolidin-1-yl)methyl]-2-chloro-benzamide, a hydrochloride salt.
  • 14. 14 Table 1. Recently tested compounds with potential antagonistic activity on P2X7 Receptor [63] Compound Study Trial Phase Completed Observed Outcome Side effects A-438079 Neuropathic and inflammatory pain Pre- clinical Yes Inhibited mechanical allodynia and effective in the formalin pain model Not evaluated. A-740003 Inflammatory and neuropathic pain Pre- clinical Yes Analgesic effect in inflammatory and neuropathic rat models Not evaluated A-804598 Neuropathic and inflammatory pain Pre- clinical Yes Analgesic effect in inflammatory and neuropathic rat models Not evaluated A847227 Inflammatory and neuropathic pain Pre- clinical Yes Antiallodynic effect and decreased IL- 1β released in mouse model Not evaluated AZ10606120 Ligand interaction and binding to P2X7R Pre- clinical Yes Had allosteric effect of receptor activity as it binds non- ATP binding site Not evaluated AZD9056 Rheumatoid Arthritis IIb Yes No significant efficacy Gastrointestinal (vomiting, nausea and diarhoea GSK314118A Inflammatory pain Pre- clinical Yes Analgesic effect in rat CFA model of inflammatory hyperalgesia Not related Natural products as novel source of analgesics for targeting ATP-mediated P2X receptors Attempts to overcome the problem of non-selectivity of P2X receptor antagonists resulted in the search for clues from natural products. Natural products have been shown to be potentially good source of new specific molecules for the
  • 15. 15 treatment of different pain syndromes. Thus, several natural products have been developed which has antagonistic properties on P2X receptors in chronic neuropathic and inflammatory pain, including Emolin, Amentoflavone, Ligunstrazine, puerarin and purotoxin-1 [64]. For instance, it was shown in rats that following formalin-induced pain, the herbal product used in Chinese medicine called, Ligunstrazine (tetramethypyrazine) derived from Ligusticum wallichii, antagonise P2X3 receptor resulting in the inhibition of membrane depolarisation induced in DRGs neurons [65]. The same group confirmed this in another study, where they also found that Ligunstrazine inhibited P2X3 receptor resulting in reduced ionic currents induced by ATP in the DRG neurons but it was non-selective since it also induces PKC activation. Similarly, in a neuropathic pain model it was also found that the Ligunstrazine inhibited the activation of P2X3 receptors on the primary afferent neurons. Furthermore, other natural products including puerarin were discovered and showed to inhibit burn-associated hyperalgesia by preventing the upregulation of P2X3 receptor expression in the DRG neurons, with similar analgesic effect on neuropathic pain via the same mechanism [64]. Additionally, Emodin, an anthraquinone obtained from rhubarb extract did not only show analgesic activity on neuropathic pain through the inhibition of P2X3 receptors in the primary sensory neurons, but also had antagonistic activity on the P2X7 receptors [64]. Also, a peptide isolated from the venom of the Asian spider Geolyscosa species, Purotoxin, has been shown to be a potent and selective antagonist of P2X3 receptors, as it selectively blocks the P2X3 receptor ion current in rat neurons [64]. Thus, these evidences collectively suggest that natural products could offer more potent and selective compounds for therapeutic targeting of P2X receptors. Conclusion Pain is the unpleasant sensory and emotional experience associated with the actual or potential tissue damage. It can simply be classified into acute and chronic pain where acute pain describes the actual pain sensation experienced in response to injury or tissue damage. Chronic pain, which can further be subdivided in inflammatory and neuropathic pain, occurs due to neurochemical and phenotypic sensitisation of the peripheral central sensory nerves characterised by increased sensitivity to painful stimuli (hyperalgesia) and the perception of pain in response to normally innocuous stimuli (allodynia). Pain sensitisation usually occurs in response to tissue damage or inflammation and is mediated by several pronociceptive neurotransmitters and neurotrophic factors such as ATP or BDNF. Extracellular ATP release has now been shown to elicit and maintain sensations following inflammation and nerve injury by activating
  • 16. 16 homomeric and heteromeric P2X receptors notably P2X2/3, P2X3, P2X4 and P2X7 receptors on the peripheral nerves and glial cells (astrocytes and microglia) in the spinal cord. Furthermore, several evidences suggest the invaluable role P2X2/3, P2X3, P2X4 and P2X7 receptors in the molecular processes involved in pain sensation, while inhibition of receptor activity using ASO, gene knockout and chemical compounds collectively showed that the absence of P2X receptors reverts the ATP-induced nociceptive pain. Thus, suggesting that inhibition of these receptors could be a potential therapeutic strategy for development of future analgesics. Massive studies in this area have resulted in the discovery of P2XR-targetted therapeutics or molecules. These P2XR antagonists include MK3901, AZ-2, A740003, AZD9056, AZ10606120, A847227, etc which have been shown to effectively block receptor activity although majority of them failed clinical trials. This is mostly due to the widespread expression of all the different receptor subtypes, which renders the discrimination between beneficial and side effects extremely complicated. For instance, studies showed that double-knockout P2X2/P2X3 receptors in the gustatory nerves of mice eliminates taste responses, although the nerve was responsive to touch, temperature and menthol [66], [67] while P2X4R-/- in mice had also been shown to cause increased blood pressure and decreased excretion of nitric oxide products in their urine compared to wildtype [68]. Similarly, the lack of good subtype-selective agents, and when selective ligands are available, the lack of an acceptable route of administration to humans are potential challenges to the development of novel P2XR targets. Other challenges include the restrictive tissue distribution of some of these receptors; the ability of channels to form heterotrimers, the ability of the extracellular domain to undergo substantial conformational rearrangement on channel opening as well as limited information on the role of P2XRs in physiological and pathological processes, although this roles are now beginning to be identified through knockout mice studies [21]. However, the good news is that crystal structures of some of these receptors are now available in the closed and ATP-bound (open) states. Hence, in the nearest future we will begin to fully understand the mechanism of action of these receptors and how to pharmacologically inhibit them. Further work could thus, explore the potential of natural products as possible source of future P2XR-targeted therapeutics as well as identifying possible molecules that could potently inhibit P2X4 receptor. Reference [1] T. Trang, S. Beggs, and M. W. Salter, “ATP receptors gate microglia signaling in neuropathic pain.,” Exp. Neurol., vol. 234, no. 2, pp. 354–61, Apr. 2012.
  • 17. 17 [2] V. Tiwari, Y. Guan, and S. N. Raja, “Modulating the delicate glial-neuronal interactions in neuropathic pain: promises and potential caveats.,” Neurosci. Biobehav. Rev., vol. 45, pp. 19–27, Sep. 2014. [3] E. Toulme, M. Tsuda, B. S. Khakh, and K. Inoue, “On the Role of ATP-Gated P2X Receptors in Acute, Inflammatory and Neuropathic Pain,” in Translational Pain Research: From Mouse to Man, CRC Press, 2010. [4] C. E. Jahr and T. M. Jessell, “ATP excites a subpopulation of rat dorsal horn neurones,” Nature, vol. 304, no. 5928, pp. 730–733, Aug. 1983. [5] P. Holton, “The liberation of adenosine triphosphate on antidromic stimulation of sensory nerves.,” J. Physiol., vol. 145, no. 3, pp. 494–504, Mar. 1959. [6] T. Bleehen and C. A. Keele, “Observations on the algogenic actions of adenosine compounds on the human blister base preparation.,” Pain, vol. 3, no. 4, pp. 367–77, Aug. 1977. [7] K. Inoue, “P2 receptors and chronic pain.,” Purinergic Signal., vol. 3, no. 1– 2, pp. 135–44, Mar. 2007. [8] C. C. Chen, A. N. Akopian, L. Sivilotti, D. Colquhoun, G. Burnstock, and J. N. Wood, “A P2X purinoceptor expressed by a subset of sensory neurons.,” Nature, vol. 377, no. 6548, pp. 428–31, Oct. 1995. [9] V. Ralevic and G. Burnstock, “Receptors for purines and pyrimidines.,” Pharmacol. Rev., vol. 50, no. 3, pp. 413–92, Sep. 1998. [10] G. Burnstock and J. N. Wood, “Purinergic receptors: their role in nociception and primary afferent neurotransmission,” Curr. Opin. Neurobiol., vol. 6, no. 4, pp. 526–532, Aug. 1996. [11] V. Souslova, P. Cesare, Y. Ding, A. N. Akopian, L. Stanfa, R. Suzuki, K. Carpenter, A. Dickenson, S. Boyce, R. Hill, D. Nebenuis-Oosthuizen, A. J. Smith, E. J. Kidd, and J. N. Wood, “Warm-coding deficits and aberrant inflammatory pain in mice lacking P2X3 receptors.,” Nature, vol. 407, no. 6807, pp. 1015–7, Oct. 2000. [12] M. F. Jarvis, E. C. Burgard, S. McGaraughty, P. Honore, K. Lynch, T. J. Brennan, A. Subieta, T. Van Biesen, J. Cartmell, B. Bianchi, W. Niforatos, K. Kage, H. Yu, J. Mikusa, C. T. Wismer, C. Z. Zhu, K. Chu, C.-H. Lee, A. O. Stewart, J. Polakowski, B. F. Cox, E. Kowaluk, M. Williams, J. Sullivan, and C. Faltynek, “A-317491, a novel potent and selective non-nucleotide antagonist of P2X3 and P2X2/3 receptors, reduces chronic inflammatory and neuropathic pain in the rat.,” Proc. Natl. Acad. Sci. U. S. A., vol. 99, no. 26, pp. 17179–84, Dec. 2002.
  • 18. 18 [13] M. Tsuda, H. Tozaki-Saitoh, and K. Inoue, “Pain and purinergic signaling.,” Brain Res. Rev., vol. 63, no. 1–2, pp. 222–32, May 2010. [14] J. Scholz and C. J. Woolf, “Can we conquer pain?,” Oct. 2002. [15] P. Honore, K. Kage, J. Mikusa, A. T. Watt, J. F. Johnston, J. R. Wyatt, C. R. Faltynek, M. F. Jarvis, and K. Lynch, “Analgesic profile of intrathecal P2X3 antisense oligonucleotide treatment in chronic inflammatory and neuropathic pain states in rats,” Pain, vol. 99, no. 1–2, pp. 11–19, Sep. 2002. [16] G. Magni and S. Ceruti, “The Purinergic System and Glial Cells: Emerging Costars in Nociception.,” Biomed Res. Int., vol. 2014, p. 495789, Jan. 2014. [17] I. P. Chessell, J. P. Hatcher, C. Bountra, A. D. Michel, J. P. Hughes, P. Green, J. Egerton, M. Murfin, J. Richardson, W. L. Peck, C. B. A. Grahames, M. A. Casula, Y. Yiangou, R. Birch, P. Anand, and G. N. Buell, “Disruption of the P2X7 purinoceptor gene abolishes chronic inflammatory and neuropathic pain.,” Pain, vol. 114, no. 3, pp. 386–96, Apr. 2005. [18] A. Surprenant and R. A. North, “Signaling at purinergic P2X receptors.,” Annu. Rev. Physiol., vol. 71, pp. 333–59, Jan. 2009. [19] S. McGaraughty, K. L. Chu, M. T. Namovic, D. L. Donnelly-Roberts, R. R. Harris, X.-F. Zhang, C.-C. Shieh, C. T. Wismer, C. Z. Zhu, D. M. Gauvin, A. C. Fabiyi, P. Honore, R. J. Gregg, M. E. Kort, D. W. Nelson, W. A. Carroll, K. Marsh, C. R. Faltynek, and M. F. Jarvis, “P2X7-related modulation of pathological nociception in rats.,” Neuroscience, vol. 146, no. 4, pp. 1817– 28, Jun. 2007. [20] J. A. M. Coull, S. Beggs, D. Boudreau, D. Boivin, M. Tsuda, K. Inoue, C. Gravel, M. W. Salter, and Y. De Koninck, “BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain.,” Nature, vol. 438, no. 7070, pp. 1017–21, Dec. 2005. [21] R. A. North and M. F. Jarvis, “P2X receptors as drug targets.,” Mol. Pharmacol., vol. 83, no. 4, pp. 759–69, Apr. 2013. [22] R. A. North, “Molecular Physiology of P2X Receptors,” pp. 1013–1067, 2002. [23] M. P. Abbracchio, G. Burnstock, A. Verkhratsky, and H. Zimmermann, “Purinergic signalling in the nervous system: an overview.,” Trends Neurosci., vol. 32, no. 1, pp. 19–29, Jan. 2009. [24] L.-H. Jiang, “P2X receptor-mediated ATP purinergic signaling in health and disease,” Cell Health Cytoskelet., pp. 83–101, 2012.
  • 19. 19 [25] C. V, U. L, S. O, C. J, C. S, and R. F, “P2X2 and P2X5 subunits define a new heteromeric receptor with P2X7-like properties.,” J Neurosci, pp. 4284–96, 2012. [26] S. Roger, P. Pelegrin, and A. Surprenant, “Facilitation of P2X7 receptor currents and membrane blebbing via constitutive and dynamic calmodulin binding.,” J. Neurosci., vol. 28, no. 25, pp. 6393–401, Jun. 2008. [27] G. Burnstock, “Purinergic P2 receptors as targets for novel analgesics.,” Pharmacol. Ther., vol. 110, no. 3, pp. 433–54, Jun. 2006. [28] G.-Y. Xu and L.-Y. M. Huang, “Peripheral Inflammation Sensitizes P2X Receptor-Mediated Responses in Rat Dorsal Root Ganglion Neurons,” J. Neurosci., vol. 22, no. 1, pp. 93–102, Jan. 2002. [29] R. A. North, “P2X3 receptors and peripheral pain mechanisms.,” J. Physiol., vol. 554, no. Pt 2, pp. 301–8, Jan. 2004. [30] S. G. Hamilton, J. Warburton, A. Bhattacharjee, J. Ward, and S. B. McMahon, “ATP in human skin elicits a dose-related pain response which is potentiated under conditions of hyperalgesia.,” Brain, vol. 123 ( Pt 6, pp. 1238–46, Jun. 2000. [31] S. G. Hamilton, S. B. McMahon, and G. R. Lewin, “Selective activation of nociceptors by P2X receptor agonists in normal and inflamed rat skin.,” J. Physiol., vol. 534, no. Pt. 2, pp. 437–45, Jul. 2001. [32] D. A. Cockayne, S. G. Hamilton, Q. M. Zhu, P. M. Dunn, Y. Zhong, S. Novakovic, A. B. Malmberg, G. Cain, A. Berson, L. Kassotakis, L. Hedley, W. G. Lachnit, G. Burnstock, S. B. McMahon, and A. P. Ford, “Urinary bladder hyporeflexia and reduced pain-related behaviour in P2X3-deficient mice.,” Nature, vol. 407, no. 6807, pp. 1011–5, Oct. 2000. [33] J. Barclay, S. Patel, G. Dorn, G. Wotherspoon, S. Moffatt, L. Eunson, S. Abdel’al, F. Natt, J. Hall, J. Winter, S. Bevan, W. Wishart, A. Fox, and P. Ganju, “Functional Downregulation of P2X3 Receptor Subunit in Rat Sensory Neurons Reveals a Significant Role in Chronic Neuropathic and Inflammatory Pain,” J. Neurosci., vol. 22, no. 18, pp. 8139–8147, Sep. 2002. [34] G.-Y. Xu, G. Li, N. Liu, and L.-Y. M. Huang, “Mechanisms underlying purinergic P2X3 receptor-mediated mechanical allodynia induced in diabetic rats.,” Mol. Pain, vol. 7, p. 60, Jan. 2011. [35] Y. Dai, T. Fukuoka, H. Wang, H. Yamanaka, K. Obata, A. Tokunaga, and K. Noguchi, “Contribution of sensitized P2X receptors in inflamed tissue to the mechanical hypersensitivity revealed by phosphorylated ERK in DRG neurons.,” Pain, vol. 108, no. 3, pp. 258–66, Apr. 2004.
  • 20. 20 [36] M. S. Ramer, E. J. Bradbury, and S. B. McMahon, “Nerve growth factor induces P2X3 expression in sensory neurons,” J. Neurochem., vol. 77, no. 3, pp. 864–875, Dec. 2001. [37] M. Tsuda, Y. Shigemoto-Mogami, S. Koizumi, A. Mizokoshi, S. Kohsaka, M. W. Salter, and K. Inoue, “P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury.,” Nature, vol. 424, no. 6950, pp. 778–83, Aug. 2003. [38] L. Ulmann, J. P. Hatcher, J. P. Hughes, S. Chaumont, P. J. Green, F. Conquet, G. N. Buell, A. J. Reeve, I. P. Chessell, and F. Rassendren, “Up-regulation of P2X4 receptors in spinal microglia after peripheral nerve injury mediates BDNF release and neuropathic pain.,” J. Neurosci., vol. 28, no. 44, pp. 11263–8, Oct. 2008. [39] K. Nasu-Tada, S. Koizumi, M. Tsuda, E. Kunifusa, and K. Inoue, “Possible involvement of increase in spinal fibronectin following peripheral nerve injury in upregulation of microglial P2X4, a key molecule for mechanical allodynia.,” Glia, vol. 53, no. 7, pp. 769–75, May 2006. [40] M. Tsuda, E. Toyomitsu, T. Komatsu, T. Masuda, E. Kunifusa, K. Nasu-Tada, S. Koizumi, K. Yamamoto, J. Ando, and K. Inoue, “Fibronectin/integrin system is involved in P2X(4) receptor upregulation in the spinal cord and neuropathic pain after nerve injury.,” Glia, vol. 56, no. 5, pp. 579–85, 2008. [41] M. Tsuda, H. Tozaki-Saitoh, T. Masuda, E. Toyomitsu, T. Tezuka, T. Yamamoto, and K. Inoue, “Lyn tyrosine kinase is required for P2X(4) receptor upregulation and neuropathic pain after peripheral nerve injury.,” Glia, vol. 56, no. 1, pp. 50–8, 2008. [42] J. A. M. Coull, D. Boudreau, K. Bachand, S. A. Prescott, F. Nault, A. Sík, P. De Koninck, and Y. De Koninck, “Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain.,” Nature, vol. 424, no. 6951, pp. 938–42, Aug. 2003. [43] T. Trang, S. Beggs, X. Wan, and M. W. Salter, “P2X4-receptor-mediated synthesis and release of brain-derived neurotrophic factor in microglia is dependent on calcium and p38-mitogen-activated protein kinase activation.,” J. Neurosci., vol. 29, no. 11, pp. 3518–28, Mar. 2009. [44] M. Tsuda, K. Kuboyama, T. Inoue, K. Nagata, H. Tozaki-Saitoh, and K. Inoue, “Behavioral phenotypes of mice lacking purinergic P2X4 receptors in acute and chronic pain assays.,” Mol. Pain, vol. 5, p. 28, Jan. 2009. [45] L. Ulmann, H. Hirbec, and F. Rassendren, “P2X4 receptors mediate PGE2 release by tissue-resident macrophages and initiate inflammatory pain.,” EMBO J., vol. 29, no. 14, pp. 2290–300, Jul. 2010.
  • 21. 21 [46] A. Colomar, V. Marty, C. Médina, C. Combe, P. Parnet, and T. Amédée, “Maturation and release of interleukin-1beta by lipopolysaccharide- primed mouse Schwann cells require the stimulation of P2X7 receptors.,” J. Biol. Chem., vol. 278, no. 33, pp. 30732–40, Aug. 2003. [47] N. Arulkumaran, R. J. Unwin, and F. W. Tam, “A potential therapeutic role for P2X7 receptor (P2X7R) antagonists in the treatment of inflammatory diseases.,” Expert Opin. Investig. Drugs, vol. 20, no. 7, pp. 897–915, Jul. 2011. [48] J. M. Labasi, N. Petrushova, C. Donovan, S. McCurdy, P. Lira, M. M. Payette, W. Brissette, J. R. Wicks, L. Audoly, and C. A. Gabel, “Absence of the P2X7 receptor alters leukocyte function and attenuates an inflammatory response.,” J. Immunol., vol. 168, no. 12, pp. 6436–45, Jun. 2002. [49] R. E. Sorge, T. Trang, R. Dorfman, S. B. Smith, S. Beggs, J. Ritchie, J.-S. Austin, D. V Zaykin, H. Vander Meulen, M. Costigan, T. A. Herbert, M. Yarkoni- Abitbul, D. Tichauer, J. Livneh, E. Gershon, M. Zheng, K. Tan, S. L. John, G. D. Slade, J. Jordan, C. J. Woolf, G. Peltz, W. Maixner, L. Diatchenko, Z. Seltzer, M. W. Salter, and J. S. Mogil, “Genetically determined P2X7 receptor pore formation regulates variability in chronic pain sensitivity.,” Nat. Med., vol. 18, no. 4, pp. 595–9, Apr. 2012. [50] G. Ito, Y. Suekawa, M. Watanabe, K. Takahashi, T. Inubushi, K. Murasaki, N. Hirose, S. Hiyama, T. Uchida, and K. Tanne, “P2X7 receptor in the trigeminal sensory nuclear complex contributes to tactile allodynia/hyperalgesia following trigeminal nerve injury.,” Eur. J. Pain, vol. 17, no. 2, pp. 185–99, Feb. 2013. [51] C. Guo, M. Masin, O. S. Qureshi, and R. D. Murrell-Lagnado, “Evidence for functional P2X4/P2X7 heteromeric receptors.,” Mol. Pharmacol., vol. 72, no. 6, pp. 1447–56, Dec. 2007. [52] G. Dell’Antonio, A. Quattrini, E. Dal Cin, A. Fulgenzi, and M. E. Ferrero, “Antinociceptive effect of a new P2Z/P2X7 antagonist, oxidized ATP, in arthritic rats,” Neurosci. Lett., vol. 327, no. 2, pp. 87–90, Jul. 2002. [53] R. J. Gum, B. Wakefield, and M. F. Jarvis, “P2X receptor antagonists for pain management: examination of binding and physicochemical properties.,” Purinergic Signal., vol. 8, no. Suppl 1, pp. 41–56, Feb. 2012. [54] S. McGaraughty, C. T. Wismer, C. Z. Zhu, J. Mikusa, P. Honore, K. L. Chu, C.-H. Lee, C. R. Faltynek, and M. F. Jarvis, “Effects of A-317491, a novel and selective P2X3/P2X2/3 receptor antagonist, on neuropathic, inflammatory and chemogenic nociception following intrathecal and intraplantar administration.,” Br. J. Pharmacol., vol. 140, no. 8, pp. 1381–8, Dec. 2003.
  • 22. 22 [55] D. Donnelly-Roberts, S. McGaraughty, C.-C. Shieh, P. Honore, and M. F. Jarvis, “Painful purinergic receptors.,” J. Pharmacol. Exp. Ther., vol. 324, no. 2, pp. 409–15, Feb. 2008. [56] E. Bohn, S. Müller, J. Lauber, R. Geffers, N. Speer, C. Spieth, J. Krejci, B. Manncke, J. Buer, A. Zell, and I. B. Autenrieth, “Gene expression patterns of epithelial cells modulated by pathogenicity factors of Yersinia enterocolitica.,” Cell. Microbiol., vol. 6, no. 2, pp. 129–41, Feb. 2004. [57] V. Hernandez-Olmos, A. Abdelrahman, A. El-Tayeb, D. Freudendahl, S. Weinhausen, and C. E. Müller, “N-substituted phenoxazine and acridone derivatives: structure-activity relationships of potent P2X4 receptor antagonists.,” J. Med. Chem., vol. 55, no. 22, pp. 9576–88, Nov. 2012. [58] T. C. Stock, B. J. Bloom, N. Wei, S. Ishaq, W. Park, X. Wang, P. Gupta, and C. A. Mebus, “Efficacy and safety of CE-224,535, an antagonist of P2X7 receptor, in treatment of patients with rheumatoid arthritis inadequately controlled by methotrexate.,” J. Rheumatol., vol. 39, no. 4, pp. 720–7, Apr. 2012. [59] E. C. Keystone, M. M. Wang, M. Layton, S. Hollis, and I. B. McInnes, “Clinical evaluation of the efficacy of the P2X7 purinergic receptor antagonist AZD9056 on the signs and symptoms of rheumatoid arthritis in patients with active disease despite treatment with methotrexate or sulphasalazine.,” Ann. Rheum. Dis., vol. 71, no. 10, pp. 1630–5, Oct. 2012. [60] L. Stokes, L.-H. Jiang, L. Alcaraz, J. Bent, K. Bowers, M. Fagura, M. Furber, M. Mortimore, M. Lawson, J. Theaker, C. Laurent, M. Braddock, and A. Surprenant, “Characterization of a selective and potent antagonist of human P2X(7) receptors, AZ11645373.,” Br. J. Pharmacol., vol. 149, no. 7, pp. 880–7, Dec. 2006. [61] P. Honore, D. Donnelly-Roberts, M. T. Namovic, G. Hsieh, C. Z. Zhu, J. P. Mikusa, G. Hernandez, C. Zhong, D. M. Gauvin, P. Chandran, R. Harris, A. P. Medrano, W. Carroll, K. Marsh, J. P. Sullivan, C. R. Faltynek, and M. F. Jarvis, “A-740003 [N-(1-{[(cyanoimino)(5-quinolinylamino) methyl]amino}-2,2- dimethylpropyl)-2-(3,4-dimethoxyphenyl)acetamide], a novel and selective P2X7 receptor antagonist, dose-dependently reduces neuropathic pain in the rat.,” J. Pharmacol. Exp. Ther., vol. 319, no. 3, pp. 1376–85, Dec. 2006. [62] K. Inoue, “The function of microglia through purinergic receptors: neuropathic pain and cytokine release.,” Pharmacol. Ther., vol. 109, no. 1– 2, pp. 210–26, Jan. 2006. [63] L. A. Alves, R. J. S. Bezerra, R. X. Faria, L. G. B. Ferreira, and V. da Silva Frutuoso, “Physiological roles and potential therapeutic applications of the P2X7 receptor in inflammation and pain.,” Molecules, vol. 18, no. 9, pp. 10953–72, Jan. 2013.
  • 23. 23 [64] R. J. Soares-Bezerra, A. S. Calheiros, N. C. da Silva Ferreira, V. da Silva Frutuoso, and L. A. Alves, “Natural Products as a Source for New Anti- Inflammatory and Analgesic Compounds through the Inhibition of Purinergic P2X Receptors.,” Pharmaceuticals (Basel)., vol. 6, no. 5, pp. 650– 8, Jan. 2013. [65] S. D. Liang, C. S. Xu, T. Zhou, H. Q. Liu, Y. Gao, and G. L. Li, “Tetramethylpyrazine inhibits ATP-activated currents in rat dorsal root ganglion neurons,” Brain Res., vol. 1040, no. 1–2, pp. 92–97, 2005. [66] T. E. Finger, V. Danilova, J. Barrows, D. L. Bartel, A. J. Vigers, L. Stone, G. Hellekant, and S. C. Kinnamon, “ATP signaling is crucial for communication from taste buds to gustatory nerves.,” Science, vol. 310, no. 5753, pp. 1495– 9, Dec. 2005. [67] A. Vandenbeuch, C. B. Anderson, J. Parnes, K. Enjyoji, S. C. Robson, T. E. Finger, and S. C. Kinnamon, “Role of the ectonucleotidase NTPDase2 in taste bud function.,” Proc. Natl. Acad. Sci. U. S. A., vol. 110, no. 36, pp. 14789–94, Sep. 2013. [68] K. Yamamoto, T. Sokabe, T. Matsumoto, K. Yoshimura, M. Shibata, N. Ohura, T. Fukuda, T. Sato, K. Sekine, S. Kato, M. Isshiki, T. Fujita, M. Kobayashi, K. Kawamura, H. Masuda, A. Kamiya, and J. Ando, “Impaired flow-dependent control of vascular tone and remodeling in P2X4-deficient mice.,” Nat. Med., vol. 12, no. 1, pp. 133–7, Jan. 2006.