animal migraine models for drug development: status and future perspectives

20
REVIEW ARTICLE Animal Migraine Models for Drug Development: Status and Future Perspectives Inger Jansen-Olesen Peer Tfelt-Hansen Jes Olesen Published online: 14 November 2013 Ó Springer International Publishing Switzerland 2013 Abstract Migraine is number seven in WHO’s list of all diseases causing disability and the third most costly neu- rological disorder in Europe. Acute attacks are treatable by highly selective drugs such as the triptans but there is still a huge unmet therapeutic need. Unfortunately, drug devel- opment for headache has almost come to a standstill partly because of a lack of valid animal models. Here we review previous models with emphasis on optimal characteristics of a future model. In addition to selection of animal spe- cies, the method of induction of migraine-like changes and the method of recording responses elicited by such mea- sures are crucial. The most naturalistic way of inducing attacks is by infusion of endogenous signaling molecules that are known to cause migraine in patients. The most valid response is recording of neural activity in the tri- geminal system. The most useful headache related responses are likely to be behavioral, allowing multiple experiments in each individual animal. Distinction is made between acute and prophylactic models and how to validate each of them. Modern insight into neurobiological mech- anisms of migraine is so good that it is only a question of resources and efforts that determine when valid models with ability to predict efficacy in migraine will be available. 1 Introduction Migraine is number seven in WHO’s list of all diseases causing disability [13] and it is the third most costly neurological disorder [4]. Even if the triptans revolution- ized the acute treatment of migraine, a huge unmet need for better or different acute treatments exists [5]. The majority of prophylactic drugs for migraine were not developed for this indication but their efficacy was discovered by seren- dipity [68]. The need for new specific prophylactic drugs is thus much higher than for acute drugs [5, 9]. In general, animal models are often of uncertain validity in predicting efficacy of new drugs. Multiple models of stroke in rodents which seemingly mimic the human condition exactly have for example shown efficacy of new drugs but this was not translated into efficacy in patients [1012]. Many other disappointments can be mentioned, but the fact remains that the pharmaceutical industry normally requires positive effects in animal models before moving novel chemical entities into clini- cal trials. Migraine drug development is currently severely hampered by a lack of generally accepted animal models with ability to predict efficacy of anti-migraine drugs. Several thorough reviews of migraine animal models have already been published [1315]. They have carefully evaluated the published literature but the scope of the present review is different. We do not review existing models one by one, but we discuss them and possible future models in relation to a number of positive characteristics of an ideal model. We focus on the following major issues: (1) type of model (2) animal species (3) methods for pro- voking migraine-like responses (4) characterization of a migraine-like response (5) how to show response to acute anti-migraine drugs and (6) how to show response to I. Jansen-Olesen Á P. Tfelt-Hansen Á J. Olesen (&) Department of Neurology, Faculty of Health and Medical Sciences, Danish Headache Center, Glostrup Hospital, University of Copenhagen, Nordre Ringvej 57, 2600 Glostrup, Denmark e-mail: [email protected] CNS Drugs (2013) 27:1049–1068 DOI 10.1007/s40263-013-0121-7

Upload: jes

Post on 21-Dec-2016

217 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Animal Migraine Models for Drug Development: Status and Future Perspectives

REVIEW ARTICLE

Animal Migraine Models for Drug Development: Statusand Future Perspectives

Inger Jansen-Olesen • Peer Tfelt-Hansen •

Jes Olesen

Published online: 14 November 2013

� Springer International Publishing Switzerland 2013

Abstract Migraine is number seven in WHO’s list of all

diseases causing disability and the third most costly neu-

rological disorder in Europe. Acute attacks are treatable by

highly selective drugs such as the triptans but there is still a

huge unmet therapeutic need. Unfortunately, drug devel-

opment for headache has almost come to a standstill partly

because of a lack of valid animal models. Here we review

previous models with emphasis on optimal characteristics

of a future model. In addition to selection of animal spe-

cies, the method of induction of migraine-like changes and

the method of recording responses elicited by such mea-

sures are crucial. The most naturalistic way of inducing

attacks is by infusion of endogenous signaling molecules

that are known to cause migraine in patients. The most

valid response is recording of neural activity in the tri-

geminal system. The most useful headache related

responses are likely to be behavioral, allowing multiple

experiments in each individual animal. Distinction is made

between acute and prophylactic models and how to validate

each of them. Modern insight into neurobiological mech-

anisms of migraine is so good that it is only a question of

resources and efforts that determine when valid models

with ability to predict efficacy in migraine will be

available.

1 Introduction

Migraine is number seven in WHO’s list of all diseases

causing disability [1–3] and it is the third most costly

neurological disorder [4]. Even if the triptans revolution-

ized the acute treatment of migraine, a huge unmet need for

better or different acute treatments exists [5]. The majority

of prophylactic drugs for migraine were not developed for

this indication but their efficacy was discovered by seren-

dipity [6–8]. The need for new specific prophylactic drugs

is thus much higher than for acute drugs [5, 9].

In general, animal models are often of uncertain

validity in predicting efficacy of new drugs. Multiple

models of stroke in rodents which seemingly mimic the

human condition exactly have for example shown efficacy

of new drugs but this was not translated into efficacy in

patients [10–12]. Many other disappointments can be

mentioned, but the fact remains that the pharmaceutical

industry normally requires positive effects in animal

models before moving novel chemical entities into clini-

cal trials. Migraine drug development is currently

severely hampered by a lack of generally accepted animal

models with ability to predict efficacy of anti-migraine

drugs.

Several thorough reviews of migraine animal models

have already been published [13–15]. They have carefully

evaluated the published literature but the scope of the

present review is different. We do not review existing

models one by one, but we discuss them and possible future

models in relation to a number of positive characteristics of

an ideal model. We focus on the following major issues:

(1) type of model (2) animal species (3) methods for pro-

voking migraine-like responses (4) characterization of a

migraine-like response (5) how to show response to acute

anti-migraine drugs and (6) how to show response to

I. Jansen-Olesen � P. Tfelt-Hansen � J. Olesen (&)

Department of Neurology, Faculty of Health and Medical

Sciences, Danish Headache Center, Glostrup Hospital,

University of Copenhagen, Nordre Ringvej 57, 2600 Glostrup,

Denmark

e-mail: [email protected]

CNS Drugs (2013) 27:1049–1068

DOI 10.1007/s40263-013-0121-7

Page 2: Animal Migraine Models for Drug Development: Status and Future Perspectives

prophylactic migraine drugs. Finally, we present a cross-

cutting discussion of all these issues.

2 Different Models have Different Virtues

Modern drug development begins with a target, usually a

receptor, an enzyme or an ion channel. Typically such

targets have been identified by academia in human or

animal experimental studies. Several novel targets for

migraine treatment have been identified and validated in

human experimental models [16]. But the industry has been

reluctant to pursue these targets. One reason is undoubtedly

the absence of evidence from animal models. Targets, such

as receptors and ion channels can be expressed in cell lines

or in other in-vitro systems allowing high throughput

screening. However, the predictive power of high

throughput is limited to the effect on the particular target

and does not reliably bespeak efficacy in migraine. Other

in-vitro models such as the study of isolated cranial blood

vessels also allow a relatively high throughput and are

somewhat closer to the disease target [17–20]. A third class

of model is in-vivo animal experimentation. This can for

example be the open or closed cranial window models to

view dural and pial artery diameter after administration of

vasoactive substances [21, 22], neurophysiological models

with recording of provoked or spontaneous responses in the

trigeminal ganglion [23] or the trigeminal nucleus caudalis

[23–26] or histological measurements of neuronal activa-

tion markers in areas involved in pain after administration

of migraine provoking agents [27, 28]. Some of these

models are rather close to migraine but the throughput is

low as every animal has to be euthanized after the exper-

iment while the number of recordings varies from one in

immunohistochemistry to several in neurophysiological

studies. The ideal animal model is obviously one that does

not require sacrificing the animal and therefore can be used

over and over again allowing crossover experiments and

evaluation of several compounds or multiple doses. Such

models would have to be behavioral. Behavioral models

have been attempted [29–31], but it remains unknown

whether rats and other animals ever experience headache

and, if so, what the behavioral correlate would be. A lot

more work therefore remains before an optimal behavioral

model has been developed and validated. Fortunately, it is

not difficult to see how such developmental work should be

done. This is discussed in the following sections.

3 Desirable Characteristics of Experimental Animals

The ideal animal model would be one with proven

migraine attacks, however; so far no natural migraine

model exists. Furthermore, we still lack the knowledge of

how to construct such a model by genetic manipulation.

For reasons of availability, generalizability and price, the

choice of species is in reality restricted to mice and rats.

Both have been used in previous studies. For many pur-

poses mice are too small and too difficult to work with.

This seems for example to be the case with the closed

cranial window model where a large percent of operated

mice were not valid for further experimentation and pre-

constriction of the dural arteries with endothelin-1 was

necessary before studying the effect of vasorelaxing agents

[32]. Most neurophysiological and other studies have also

been done in rats for practical purposes. Mice are however,

attractive because of the possibility of using genetically

modified animals. In fact genetically modified mice

expressing a CACNA1A gene mutation of familial hemi-

plegic migraine type 1 (FHM1) have been developed [33,

34]. The R192Q mutation of the CACNA1A causes a mild

form of FHM1 while the S218L mutation is more severe

and often lethal. These models have been used in several

physiological and behavioral experiments with consider-

able success [35]. In R192Q mutated mice a reduced

threshold and increased velocity of cortical spreading

depression (CSD) has been found [34]. Furthermore, the

model show enhanced synaptic acetylcholine release under

conditions in which the synaptic Ca2? sensors are not

saturated [34]. It also seems that the R192Q mutation has

an effect on the trigeminal system as calcitonin gene-

related peptide (CGRP) immunoreactivity was decreased in

thoracic ganglia and in the superficial lamina of the trige-

mino cervical complex in R192Q knock in mice [36]. A

major problem with the CACNA1A knock in mice seems

to be the validity of these animals for the prevalent types of

migraine, migraine without aura (MO) and migraine with

typical aura (MA). In humans it has been shown that FHM

differs from MA and MO not only in genetics but also in

the response to migraine provoking agents [37–39]. Per-

haps the FHM mice are not useful for the prevalent types of

migraine. CGRP release from the trigeminal ganglion was

increased in R192Q knock in mice as compared to wild

type mice [40]. No change in CGRP release was found in

dura mater between mutated and wild type mice [40].

During restraint these animals seem to spontaneously

express changes in facial expression that were reversed by

rizatriptan [30]. Another type of genetically modified mice

has elevated expression of human receptor activity-modi-

fying protein 1 (hRAMP1), a subunit of the CGRP receptor

[41]. These transgenic mice display light-aversive behavior

that is greatly enhanced by intracerebroventricular injec-

tion of CGRP and blocked by co-administration of the

CGRP receptor antagonist olcegepant [42]. It is still

unknown if migraine triggering substances other than

CGRP are able to enhance the light aversive behavior in

1050 I. Jansen-Olesen et al.

Page 3: Animal Migraine Models for Drug Development: Status and Future Perspectives

these mice. Thus, validity of this model for the develop-

ment of new anti-migraine substances is still unknown.

Recently, a rat exhibiting episodes of spontaneous tri-

geminal allodynia (STA) has been presented as the spon-

taneous trigeminal allodynia (STA) rat model of primary

headache [43]. This STA rat has five features in common

with migraineurs: (1) Episodically changing trigeminal

thresholds to von Frey filaments, possibly reflecting tri-

geminal hypersensitivity. (2) The STA trait is inherited. (3)

The STA rats have increased sensitivity to sound as com-

pared to normal rats. (4) Increased sensitivity to the

headache triggers GTN and CGRP. (5) The STA rats

respond to commonly used acute and preventive migraine

therapeutics [43]. The model is interesting in the sense that

it is spontaneous rather than provoked and does not need

any manipulations in order to show the desired phenotype.

To further validate and increase face-and construct validity

of the STA model it must be shown that it is in fact having

episodes of spontaneous migraine and not just allodynia.

This can be done through testing of spontaneous behaviors

detecting clinical manifestations of headache. One major

disadvantage using a spontaneous model compared to a

provocation model is the episodic and unpredictable nature

of allodynia. Therefore the animals need to be tested every

day in order to determine whether they have allodynia

or not.

Male animals have generally been preferred in previous

migraine models to avoid variability of the female cycle.

However, migraine patients are mostly female and

migraine drugs are expected to work throughout the cycle.

Future migraine models should therefore explore the use of

female animals and the role of menstrual cycle on the

effects explored. Animals should be in the reproductive age

but relatively young as a mimic of the human situation. As

migraines do not occur as frequently during a stable low or

a stable high concentration of estrogen, we suggest that

experiments should be performed in animals that are not

ovariectomized and not treated with estrogen. However,

the status of the hormonal cycle could be investigated

during or immediately after experimentation.

4 Inducing a Migraine-like Response in Animals

A number of procedures have been used to induce a

response that may be related to migraine. The so-called

neurogenic inflammation model was among the first animal

models of migraine [44, 45]. Strong electrical stimulation

is applied to the trigeminal ganglion (0.6–3.0 mA with

square pulses of 5 ms duration for 5 min) [46, 47]. This

results in retrograde activity in trigeminal nerve fibers and

liberation of a number of signaling molecules that increase

permeability for plasma protein of the vessels in the dura

mater [46]. The consequent increase in water content can

easily be recorded with 125I-BSA or Evans blue. This

model has been extensively characterized in a series of

excellent experimental studies (Fig. 1; Table 1). For some

time it looked promising in relation to drug development

for migraine as drugs for migraine treatment were effective

in blocking this response (Fig. 1) [47–51]. Unfortunately, it

turned out that many drugs work in this model but have

absolutely no efficacy in migraine [52–59] (Table 1). It

remains possible, however, that this model is sensitive to

acute antimigraine drugs albeit not specific.

In the genuine closed cranial window model the dural

artery is dilated by release of CGRP evoked by a much

milder electrical stimulation (50–300 lA for 10 s). Using

this model a more close relation to the clinical situation is

found, as neurokinin1 (NK1) receptor antagonists and the

endothelin antagonist bosentan, ineffective in migraine, do

not inhibit dural artery dilatation due to electrical stimu-

lation or CGRP release (Fig. 2) [60, 61]. Drugs effective in

the acute treatment of migraine such as sumatriptan, the

CGRP receptor antagonists CGRP8–37 and olcegepant and

dihydroergotamine cause inhibition [60–62] (Fig. 2;

Table 2).

Another classical inducing procedure is cortical

spreading depression (CSD) [63]. It is more and more

likely that CSD is the mechanism underlying the migraine

aura [64]. Furthermore, a drug developed for its ability to

inhibit cortical spreading depression, tonabersat (Fig. 3)

[65], has prophylactic efficacy against migraine with aura

Fig. 1 The effect of sumatriptan on plasma extravasation produced in

the dura mater of the rat by electrical stimulation (0.6 mA, 5 ms,

5 Hz for 5 min) of the right trigeminal ganglion. Data is expressed as

the ratio of extravasation on the stimulated/unstimulated sides.

Histograms show effects of pretreatment (i.v.) with vehicle (control,

distilled H2O, n = 10), sumatriptan (1–1,000 lg/kg, n = 9–10).

Statistical analysis was performed by unpaired t-test; *P \ 0.05 from

vehicle, bars SEM (adapted from Shepheard et al. [59])

Animal Migraine Models 1051

Page 4: Animal Migraine Models for Drug Development: Status and Future Perspectives

Table 1 Neurogenic

inflammation model: effect of

different inhibitors on dural

plasma protein extravasation

Treatment Dose (i.v) Inhibition of dural

plasma protein

extravasation

References

Electrical stimulation 1.2 mA Indomethacin 1 mg/kg ?? [50]

2 mg/kg ??

Acetylsalicylic acid 10 mg/kg ?

50 mg/kg ??

Dexamethasone 1 mg/kg ?

Electrical stimulation 1.2 mA Sumatriptan 30 lg/kg – [51]

100 lg/kg ???

300 lg/kg ?

Electrical stimulation 1.2 mA a-methylhistamine 5 lmol/kg – [165]

15 lmol/kg ???

SMS 2111-905 (somatostatin agonist) 0.1 lmol/kg –

0.3 lmol/kg –

1 lmol/kg ???

UK-14,304 (a2 receptor agonist) 34 nmol/kg –

100 nmol/kg ?

340 nmol/kg ??

Electrical stimulation 1.2 mA Sodium valproate 3 mg/kg ? [49]

10 mg/kg ??

30 mg/kg ??

100 mg/kg ??

Electrical stimulation 1.2 mA Muscimol 10 lg/kg ? [49]

100 lg/kg ??

1,000 lg/kg ??

Baclofen 10 lg/kg –

100 lg/kg –

1 mg/kg –

10 mg/kg –

Electrical stimulation 1.2 mA GR82334 (NK1 receptor antagonist) 0.02 mg/kg ? [166]

0.2 mg/kg ?

SR 48968 (NK2 receptor antagonist) 1 mg/kg –

CGRP (8–37) 0.1 mg/kg –

Electrical stimulation 1.2 mA Bosentan (mixed ET receptor

antagonist)

10 mg/kg ?? [52]

30 mg/kg ??

BQ-123 (ETA receptor antagonist) 10 mg/kg –

Ro-46-8443 (ETB receptor antagonist) 10 mg/kg ???

Electrical stimulation 1.2 mA Sumatriptan 1 lg/kg – [167]

10 lg/kg –

100 lg/kg ?

1 mg/kg ?

CP 122,288 (5-HT1B/1D receptor

agonist)

1 pg/kg –

10 pg/kg –

100 pg/kg ?

10 lg/kg –

100 lg/kg –

CP 93,129 (5-HT1B/1D receptor

agonist)

1 mg/kg ?

3 mg/kg ?

Electrical stimulation 1.2 mA 100 % oxygen 200 mmHg ? [47]

300 mmHg ??

400 mmHg ??

1052 I. Jansen-Olesen et al.

Page 5: Animal Migraine Models for Drug Development: Status and Future Perspectives

Table 1 continuedTreatment Dose (i.v) Inhibition of

dural

plasma protein

extravasation

References

Electrical stimulation 0.6 mA CP 99,994 (NK1 receptor antagonist) 10 lg/kg – [59]

100 lg/kg ?

1 mg/kg ?

3 mg/kg ?

Sumatriptan 10 lg/kg –

100 lg/kg ?

1 mg/kg ?

Electrical stimulation 0.6 mA Rizatriptan 1 lg/kg – [136]

10 lg/kg –

100 lg/kg ?

1,000 lg/kg ?

Electrical stimulation 1.2 mA (guinea

pig)

SMS 2111–905 (somatostatin agonist) 0.1 lmol/kg – [165]

0.3 lmol/kg ?

1 lmol/kg ???

Electrical stimulation 1.5 mA (guinea

pig)

GR82334 (NK1 receptor antagonist) 0.02 mg/kg – [166]

0.2 mg/kg ?

CGRP (8–37) 0.1 mg/kg ?

Electrical stimulation 1.5 mA (guinea

pig)

PNU 10929 (5-HT1D agonist) 0.24 nmol/

kg

– [168]

2.4 nmol/kg ??

7.3 nmol/kg ??

24.4 nmol/

kg

??

73.3 nmol/

kg

??

Substance P (1 nmol/kg) Indomethacin 2 mg/kg – [50]

10 mg/kg ?

Acetylsalicylic acid 10 mg/kg –

50 mg/kg ?

Dexamethasone 1 mg/kg –

Substance P (1 nmol/kg) Sodium valproate 3 mg/kg ? [49]

10 mg/kg ?

30 mg/kg ?

100 mg/kg ?

Muscimol 0.3 mg/kg ?

1 mg/kg ?

30 mg/kg ?

Substance P (1 nmol/kg) Sumatriptan 100 lg/kg – [51]

Substance P (0.3 nmol/kg) Sumatriptan 100 lg/kg – [51]

300 lg/kg –

Bradykinin (0.1 lmol/kg) Sumatriptan 10 lg/kg – [51]

30 lg/kg ?

100 lg/kg ?

Capsaicin (1 lmol/kg) Sumatriptan 30 lg/kg – [51]

100 lg/kg ?

Capsaicin (0.5 lmol/kg) (guinea pig) Sumatriptan 10 lg/kg – [51]

30 lg/kg ?

Capsaicin (1 lmol/kg) a-Methylhistamine (histamine H3

receptor agonist)

15 lmol/kg ?? [165]

SMS 201–995 (somatostatin

analogue)

1 lmol/kg ???

UK-14,304 (alpha-adrenoceptor

agonist)

100 nmol/kg ??

Capsaicin (0.37 mg/kg) Bosentan (mixed ET receptor

antagonist)

10 mg/kg ?? [52]

30 mg/kg

Sumatriptan 300 lg/kg ??

Animal Migraine Models 1053

Page 6: Animal Migraine Models for Drug Development: Status and Future Perspectives

[66], but not against migraine without aura [67]. Taken

together with extensive human brain blood flow studies it

seems overwhelmingly likely that cortical spreading

depression in animals is a valid model for the testing of

prophylactic drugs against migraine with aura [66, 68]. The

number of patients with a high attack frequency of

migraine with aura is, however, limited. Probably the small

market size for this indication is the reason why the pharma

industry has made little use of the cortical spreading

depression model for drug development. Whether this

model might also be relevant to migraine without aura is

debatable. From human brain blood flow studies there is no

indication of CSD in migraine without aura [69, 70], but

prophylactic migraine drugs seem to inhibit CSD in rats

when dosed for two weeks or more [71, 72]. Thus, CSD

models may perhaps predict efficacy of prophylactic drugs

not only for migraine with aura but also for migraine

without aura.

Other models use stimulation of cranial vascular structures,

primarily the sagittal sinus [24, 73–79] but also the middle

meningeal artery [80, 81] (Table 3). This is associated with

activation of areas of the central nervous system that are rel-

evant to headache perception. Thus, it induces c-Fos expres-

sion in the nucleus caudalis [24, 75, 80–83] and activation of

neurophysiological responses [77, 79, 81, 84]. This model

responds to some anti-migraine drugs (Fig. 4) [24, 77, 78, 82,

83, 85] and failed to respond to NK1 antagonists [86] and

5-HT1D receptor specific triptans [82, 87]. These drugs are

proven ineffective in migraine. Some models have used a so-

called inflammatory soup on the dura mater in acute studies

with recording of neural responses in the pain pathway [81, 88,

89]. A model was recently developed where the inflammatory

soup or other agents can be delivered supradurally to awake

freely moving rats [90]. However, it was not investigated if

this model responds to specific anti-migraine treatments.

Others have used more continuous stimulation of the dura

Fig. 2 Example traces showing

the effects of a the NK1 receptor

antagonist RP67580 (1 mg/kg,

iv) and b the CGRP receptor

antagonist human-aCGRP8–37

(0.3 mg/kg, iv) on the increase

in dural vessel diameter

produced by substance P (SP;

100 ng/kg, iv) or electrical

stimulation (ES; 50–300 lA,

5 Hz, 1 ms for 10 s) of the

cranial window. Upper trace is

blood pressure (mmHg) and

lower trace is dural vessel

diameter (arbitrary units)

(adapted from Williamson et al.

[22])

Table 1 continued

Significant effect ? P \ 0.05,?? P \ 0.01, ??? P \ 0.001

Treatment Dose (i.v) Inhibition of dural

plasma protein

extravasation

References

Substance P (1 nmol/kg) a-Methylhistamine (histamine H3

receptor agonist)

15 lmol/kg – [165]

SMS 201–995 (somatostatin

analogue)

1 lmol/kg –

UK-14,304 (alpha-adrenoceptor

agonist)

100 nmol/kg –

GTN (60 lg/kg iv) L-NMMA 20 mg/kg iv ? [113]

L-NIL (iNOS inhibitor) 4 mg/kg ip ?

1054 I. Jansen-Olesen et al.

Page 7: Animal Migraine Models for Drug Development: Status and Future Perspectives

Table 2 The genuine closed cranial window model: effect of dif-

ferent inhibitors on middle meningeal artery (MMA) diameter change

after transcranial electrical stimulation

Treatment Dose (i.v) Inhibition

of arterial

dilatation

References

Olcegepant (CGRP

antagonist)

3 lg/kg – [62]

10 lg/kg ?

30 lg/kg ?

100 lg/kg ?

300 lg/kg ?

Iberiotoxin (BKCa

channel blocker)

0.1 mg/kg – [169]

NOX-C89 (CGRP

binding Spiegelmer)

1 mg/kg – [170]

CGRP antibody 10 mg/kg –

Glibenclamide (KATP

channel blocker)

7 mg/kg ? [171]

20 mg/kg ??

30 mg/kg ??

Ketamine (NMDA

receptor antagonist)

10 mg/kg – [172]

18 mg/kg ?

30 mg/kg ?

MK801 (NMDA receptor

antagonist)

0.5 mg/kg –

1 mg/kg –

3 mg/kg ?

GYKI52466 (AMPA

receptor antagonist)

0.5 mg/kg –

2 mg/kg –

5 mg/kg –

LY466195 (kainate

receptor antagonist)

0.03 mg/kg –

0.1 mg/kg –

0.3 mg/kg –

Sumatriptan 1 mg/kg – [60]

3 mg/kg ?

10 mg/kg ?

RP67580 (NK1 receptor

antagonist)

1 mg/kg –

CGRP (8–37) 0.3 mg/kg ?

Rizatriptan 1 mg/kg – [136]

3 mg/kg ?

10 mg/kg ?

Nociceptin 1 nmol/kg ?? [173]

10 nmol/kg ??

100 nmol/kg ??

L-NAME 40 mg/kg ? [174]

Diphenylene-iodonium

(eNOS inhibitor)

0.1 mg/kg –

0.3 mg/kg –

SMTC (nNOS inhibitor) 1 mg/kg –

3 mg/kg –

10 mg/kg ?

SMT (iNOS inhibitor) 3 mg/kg –

10 mg/kg –

Table 2 continued

Treatment Dose (i.v) Inhibition

of arterial

dilatation

References

Sumatriptan 1 mg/kg – [167]

10 mg/kg ?

CP 122,288 (5-HT1B/1D

receptor agonist)

1 mg/kg –

10 mg/kg ?

CP 93,129 (5-HT1B

receptor agonist)

1 lg/kg –

10 lg/kg ?

100 lg/kg ?

1 mg/kg ?

Flunarizine 1 mg/kg – [175]

2.5 mg/kg –

Indomethacin 3 mg/kg –

10 mg/kg ?

Phenylephrine

(a1-adrenoceptor

agonist)

1 lg/kg – [176]

5 lg/kg –

Corynanthine (a1-

adrenoceptor

antagonist)

1 mg/kg –

UK 14,304

(a2-adrenoceptor

agonist)

2 mg/kg –

Yohimbine (a2-

adrenoceptor

antagonist)

5 lg/kg –

10 lg/kg –

1 mg/kg –

3 mg/kg –

Propranolol (b-

adrenoceptor

antagonist)

1 mg/kg –

3 mg/kg –

Mepyramine (H1-receptor

antagonist)

1 mg/kg – [177]

3 mg/kg –

10 mg/kg ?

Famotidine (H2 receptor

antagonist)

1 mg/kg –

3 mg/kg –

10 mg/kg –

Calciseptine (L-type

voltage dependent

calcium channel

blocker)

7 lg/kg – [178]

10 lg/kg –

20 lg/kg ?

x-Agatoxin-TK (P/Q-

type voltage-dependent

calcium channel

blocker)

3 lg/kg –

10 lg/kg ?

20 lg/kg ?

x-Agatoxin-IVA (P/Q-

type voltage-dependent

calcium channel

blocker)

3 lg/kg ?

10 lg/kg ?

20 lg/kg ?

x-Conotoxin-GVIA (N-

type voltage-dependent

calcium channel

blocker)

10 lg/kg ?

20 lg/kg ?

40 lg/kg ?

Animal Migraine Models 1055

Page 8: Animal Migraine Models for Drug Development: Status and Future Perspectives

mater for example with endotoxin resulting in a chronic

inflammation [29]. This model has responded to anti-migraine

drugs but the relevance of such strong inflammation to

migraine is otherwise improbable. Pain stimulation in the extra

cranial facial tissues such as injection of nociceptive agents in

the jaw joint or injection into chewing muscles of formalde-

hyde have also been used [91]. Since migraine patients do not

normally encounter pain in facial structures, this mode of

stimulation may not be close enough to migraine pathology.

5 Pharmacological Migraine Provocation in Man

and Animal

In relation to behavioral animal models it may be an

advantage to mimic human experimental models. Such

models have been developed over the last 20 years and

have been extensively characterized in normal individuals

and migraine sufferers. A number of naturally occurring

signaling substances or drugs interacting with known

pathological pathways are able to induce headache in

normal volunteers and migraine attacks in migraine suf-

ferers [92–102]. These human models should be further

developed to be practical in the testing of new drugs. But,

they will never replace the need for animal models.

However, it should be noted that the translational value of

such animal models is diminished by the differences

between migraine sufferers and normal animals. The most

extensively studied human model uses nitroglycerin,

glyceryl trinitrate (GTN) [92, 93, 96]. It has been mim-

icked in numerous animal experimental studies (Table 4).

In rats it is believed that increase in Fos expression within

the dorsolateral lamina 1 and 2 of the caudal region of the

trigeminal nucleus caudalis (TNC) may indicate activation

of the trigeminal vascular system [103, 104]. A dose

approximately 1,000 times the human dose has unfortu-

nately been used in most animal studies [28, 105–111]. It

causes depression of blood pressure and activates c-Fos

also in areas of the brain that are not related to pain [28,

109]. Other variations of this model have used anaesthe-

tized animals [112–116]. Anesthesia and surgery by

themselves affect the expression of c-Fos [117, 118]. In

anaesthetized rats GTN-induced hypotension which by

itself causes c-Fos expression [119]. Therefore, c-Fos

expression has been confounded by a number of unspecific

factors, which indicate a major deviation from studies in

awake human subjects. A more naturalistic model in the rat

has recently been presented [27]. GTN was administered to

awake freely moving rats in a dose (4 lg/kg/min for 20

min) 8 times the human dose, probably the equivalent of

Fig. 3 Box plot of number of cortical extracellular field potential

depolarisations. Four experimental groups were utilised; vehicle

(n = 3) (methylcellulose 1 ml/kg i.p. ? saline 1 ml/kg i.v.), suma-

triptan (n = 3) (300 lg/kg i.v.), tonabersat (n = 3) (10 mg/kg i.p.)

and sham (n = 3). Sham experiments included all surgery excepting

initiation of CSD. Data represented as medians and 25–75 % range.

Significant differences between groups were analysed by Kruskal–

Wallis and Mann–Whitney U test, **P \ 0.01 versus vehicle

(adapted from Read et al. [160])

Table 2 continued

Treatment Dose (i.v) Inhibition

of arterial

dilatation

References

LY334370 (5-HT1F

receptor agonist)

3 mg/kg – [179]

10 mg/kg –

Morphine 100 lg/kg – [180]

1 mg/kg ?

DAGO (l-opioid receptor

antagonist)

1 lg/kg ?

10 lg/kg ?

100 lg/kg ?

Butorphanol 1 mg/kg ?

10 mg/kg ?

DPDPE (j-opioid

receptor antagonist)

1 mg/kg –

U 50,488 (d-opioid

receptor antagonist)

100 lg/kg –

Orexin A 3 lg/kg – [181]

10 lg/kg –

30 lg/kg ?

Orexin B 3 lg/kg –

10 lg/kg –

30 lg/kg –

Anandamide 1 mg/kg ? [182]

3 mg/kg ?

Topiramate 10 mg/kg – [183]

30 mg/kg ?

GR79236 (adenosine A1

receptor antagonist)

1 lg/kg – [184]

3 lg/kg –

10 lg/kg ??

Significant effect ? P \ 0.05, ?? P \ 0.01, ??? P \ 0.001

1056 I. Jansen-Olesen et al.

Page 9: Animal Migraine Models for Drug Development: Status and Future Perspectives

the human dose in rats. A significant increase in c-Fos

mRNA expression was observed in the trigeminal nucleus

caudalis at 30 min and 2 h that was followed by an

increase in Fos protein in the trigeminal nucleus caudalis at

2 and 4 h after GTN infusion (Fig. 5). Treatment with

sumatriptan and non-selective NOS inhibitor L-NAME as

well as pre-treatment with the CGRP receptor antagonist

olcegepant attenuated the activation of c-Fos at 4 h

Table 3 Stimulation of cat superior sagittal sinus: the table shows different effects observed after electrical stimulation of the superior sagittal

sinus and the effect of different treatments on these effects

Effect observed Treatment Effect studied by

treatment

References

Electrical stimulation of SSS

0.3 Hz 120 min

3 Hz 45 min

Increase of c-Fos in laminae I/II ofTNC, C1–C3

No treatment [185]

Electrical stimulation

0.3 Hz 120 min

Increase of c-Fos in laminae I/II of

TNC, C1–C2

L-NAME, 100 mg/kg ? [83]

Electrical stimulation

0.3 Hz 120 min

Increase of c-Fos in laminae I/II of

TNC, C1–C2

Eletriptan, 100 ng/kg – [82]

CP122,288, 0.5 mg/kg (5-HT1B/1D

receptor agonist)?

Electrical stimulation of SSS

150 V, 250 ls duration, 0.3 Hz

Single unit activity in dorsolateral C2

spinal cord.

Sumatriptan – [85]

Sumatriptan ? mannitol

(disruption of BBB)

?

Electrical stimulation of SSS

150 V, 250 ls duration, 0.3 Hz

Single unit activity in dorsolateral C2spinal cord

Zolmitriptan [82]

30 lg/kg ?

100 lg/kg ?

Electrical stimulation of SSS

150 V, 500 ls duration, 10 Hz

Release of vaso-active peptides into

external jugular vein

[186]

CGRP Intact TG ?

SP –

VIP ?

NPY –

CGRP Bilateral trigeminal ablation –

SP –

VIP –

NPY –

Electrical stimulation of SSS Release of CGRP into external

jugular vein

Avitriptan (50 lg/kg) ? [187]

150 V, 500 ls duration, 10 Hz CP122, 288 (0.1 lg/kg) –

Release of CGRP into externaljugular vein

4991W93 (Zolmitriptananalogue)

[87]

0.1 lg/kg –

10 lg/kg –

100 lg/kg ?

Electrical stimulation of SSS Square wave pulsesevery 3 s, 130–150 V for 2 h

c-Fos positive neurons in: No treatment [188]

Rostral hypothalamus

Fornix –

Lat hypothalamic n. –

Ventromedial/ant. hypothalamus –

Supraoptic ?

Optic tract –

Paraventricular hypothalamus –

Caudal hypothalamus

Lat. hypothalamic n. –

Post. hypothalamus ??

Lat. mamillary –

Optic tract –

Supramamillary decussation –

Significant effect ? P \ 0.05, ?? P \ 0.01, ??? P \ 0.001

Animal Migraine Models 1057

Page 10: Animal Migraine Models for Drug Development: Status and Future Perspectives

(Fig. 5). However; a NK1 receptor antagonist that has no

efficacy in migraine also attenuated c-Fos expression [27].

Other migraine provoking agents such as CGRP, PACAP,

histamine, prostanoids and phosphodiesterase (PDE)

inhibitors have received limited or no study in this model.

Finally, it may be considered whether animals should be

sensitized in some way. GTN induces migraine in migraine

sufferers and a migraine-like relatively mild immediate

headache in normal volunteers [96]. If normal volunteers

are pre-treated for example with acetazolamide and then

receive GTN, roughly one fourth of the subjects develop a

migraine-like attack [120]. If a more reliable and stronger

sensitizing procedure could be developed, it might be

possible to induce a migraine in many more normal indi-

viduals. Perhaps sensitizing procedures could be developed

so that normal rats develop a migraine-like attack. Repe-

ated application of an inflammatory soup on the dura mater

produced a chronic state of trigeminal hypersensitivity that

potentiated the GTN evoked glutamate release in TNC

[121]. Daily treatment with sumatriptan over seven days

likewise caused allodynia that lasted for up to 14 days after

termination of sumatriptan exposure (Fig. 6) [31].

6 Recording the Response to Provocative Procedures

The responses to migraine provocation should obviously be

as close as possible to those occurring during a migraine

attack in humans. Pain is the dominant migraine symptom

and activation of the trigeminal afferent pain pathway is

therefore the most valid response. Neurophysiological

recordings in the trigeminal ganglion [122, 123] or the

trigeminal nucleus [85, 89, 123–125] or perhaps in even

higher centers in thalamus [124, 126, 127] or somatosen-

sory cortex can quantify such responses.

Activity in non-nociceptive afferent pathways can con-

found the picture and the methodology requires expertise.

The expression of c-Fos is often used as an indirect marker

of recent neuronal activity [128–130]. The up-regulated

protein expression of Fos and other biochemical markers

such as ERK, CREB and Jun reflect nociception if they are

located to areas involved in pain transmission [131, 132]. It

can be visualized by histological methods and has the

virtue of anatomical detail but the validity is not as high as

neurophysiological recordings. Furthermore, only one

measurement can be made in each animal using immuno-

histochemistry or other anatomical methods.

Extravasation in the dura mater, as in the neurogenic

inflammation model, is another possible way of recording a

response. As discussed above this may be sensitive but it is

certainly not specific to migraine. Increased dural and pial

arterial diameter or increased brain blood flow has been

used as a migraine-like response and in a human MR study

of 19 migraine patients during spontaneous migraine

attacks a slight increase in middle cerebral artery diameter,

but not of middle meningeal and extra cranial arteries were

found [133]. However, the importance of vasodilatation in

migraine has been under attack and been suggested to be an

epiphenomenon [134, 135]. However, some substances of

proven efficacy in the acute treatment of migraine such as

the triptans and olcegepant inhibit dural artery vasodilata-

tion due to electrical stimulation [60, 62, 136] (Table 2),

while a NK1 receptor antagonist that is not effective in

migraine treatment does not [61] (Table 2). For special

purposes liberation of CGRP from dura mater or other

migraine relevant tissues can be used as an outcome

parameter when testing drugs that target CGRP related

mechanism [60, 137, 138].

Recording behavioral responses to migraine provoking

stimuli is a whole issue of its own in need of much more

developmental work. However, a number of results have

already been obtained. In the genetically modified mouse

expressing a FHM1 mutation, a particular and quite

detailed facial expression that responds to a triptan has

been described to occur spontaneously [30].

Cutaneous allodynia is often present in migraineurs

[139–141] and has been induced in animals by direct

application of inflammatory mediators (inflammatory soup,

lipopolysaccharide, TNF-alpha) to the dura mater [29, 121,

142, 143]. It can also be induced by long term systemic

treatment with triptans [31]. The inflammatory response

causes increased sensibility of primary sensory nerve fibers

in dura mater which then respond to stimuli that under

normal condition would not cause activation [122]. Out-

comes were increased firing of primary or secondary tri-

geminal neurons [89, 122, 144], cutaneous allodynia

Fig. 4 Population effect of zolmitriptan on trigeminal evoked

potentials due to stimulation of SSS. The histogram illustrates the

trigeminal potential in the caudal trigeminal nucleus complex before

(control) and after 30 lg/kg (n = 5) or 100 lg (n = 4) of zolmitrip-

tan. There is a significant (* = p \ 0.05) effect on the evoked

potential at both doses. The ordinate is in lV (adapted from Goadsby

and Hoskin [74])

1058 I. Jansen-Olesen et al.

Page 11: Animal Migraine Models for Drug Development: Status and Future Perspectives

Table 4 Glyceryltrinitrate infusion studies: the table shows effects observed after infusion of GTN and the effect of different treatments on these

effects

Effect observed Treatment Inhibition of effect

studied by treatment

References

GTN 0.25 lg/kg/

min i.v. (cat)

Increase in pial artery diameter

Increase in rCBF

[114]

Anesthetized Increase in NO concentration

GTN 60 lg/kg over

30 min iv

Increase in NO concentration Sumatriptan ? [115]

Anesthetized Decrease in superoxide concentration Sumatriptan ?

Increase in rCBF Sumatriptan ?

GTN 10 mg/kg s.c. Increase in TNC c-Fos expression [107]

Increase in TNC nNOS expression

GTN 10 mg/kg s.c. Decrease in TNC CGRP-IR expression [189]

Increase in TNC 5-HT-IR expression

GTN 10 mg/kg s.c. Increase in CamKII-IR in TNC – [190]

GTN 10 mg/kg i.p. Increase in TNC c-Fos expression Kynurenin (450 mg/kg) and

probenecid (200 mg/kg)

? [191]

GTN 10 mg/kg i.p. Increase in TNC c-Fos expression Kynurenic acid (1 mmol/kg i.p.) ??? [192]

SZR-72 (kynurenate analog)

(1 mmol/kg i.p.)

???

GTN 10 mg/kg i.p. Increase in c-Fos expression in TNC and

10 other brain nuclei

[28]

GTN 10 mg/kg i.p. Increase in c-Fos expression L-NAME (50 mg/kg i.p.) ? [110]

7-NI (20 mg/kg i.p.) ?

Ephedrine (25 mg/kg ip) ?

Indomethacin (5 mg/kg ip) ?

Capsaicin depletion ?

GTN 10 mg/kg i.p. Increase in cGMP in TNC lamina I/II [193]

GTN 10 mg/kg ip Increase in c-Fos expression Parthenolide

15 mg/kg for 6 days

? [194]

GTN 10 mg/kg ip Increase in c-Fos expression Anandamide

20 mg/kg ip

? [195]

GTN 10 mg/kg sc Increase in CamKII expression in TNC NS398 (COX-2 inhibitor) [196]

1 mg/kg –

3 mg/kg ?

5 mg/kg ??

SC 560 (COX-1 inhibitor)

1 mg/kg –

3 mg/kg –

5 mg/kg –

GTN 80 lg/kg over

20 min iv

Awake rats

Increase in c-Fos expression Sumatriptan

1.8 mg/kg iv

? [27]

GTN 80 lg/kg over

20 min iv

Awake rats

Increase in c-Fos expression L-NAME

40 mg/kg iv

? [157]

Olcegepant

1 mg/kg iv

?

L-733060 (NK1 receptor

antagonist)

1 mg/kg iv

?

Animal Migraine Models 1059

Page 12: Animal Migraine Models for Drug Development: Status and Future Perspectives

located to the periorbital region of the head or to fore- or

hind paws [121, 142] and vocalization due to an air current

focused on the head of the rat [29]. While this certainly

indicates activation of the nociceptive system, the speci-

ficity to migraine is somewhat uncertain. Allodynia has

been recorded in mice after GTN challenge but with very

high doses (5–10 mg/kg) of GTN [105]. In humans without

migraine there was no change in thermal pain thresholds

and minor change in pressure pain thresholds after GTN

[145].

The migraine attack is associated with hypersensitivity

to light and sound and sometimes hypersensitivity to other

external stimuli. The amount of locomotion and grooming

of the head therefore seem relevant. Presence of photo-

phobia has been elegantly evaluated in an experimental

maze where one compartment was illuminated and animals

after GTN and CGRP avoided the light [42, 146]. The same

was demonstrated in the genetically modified ‘‘migraine’’

mouse [42]. Phonophobia has not yet been studied.

Migraine patients are also nauseated and anorexic. The

amount of food and water intake could thus be another

important parameter in a behavioural model of migraine.

Osmophobia is a specific but not very sensitive migraine

symptom [147–149]. Osmophobia could also be tested in

an animal maze. Migraine patients avoid physical activity

during attack [150]. Running wheel activity could possibly

be used as a marker of this. Little systematic work has been

done to validate different behaviors as migraine outcome

parameters. Hopefully this will happen in the near future.

7 Validating Models of Acute Migraine Treatment

We have already discussed a number of characteristics that

support the validity of experimental migraine models. The

final proof is, however, whether models respond to specific

acute anti-migraine drugs. Fortunately, there are now sev-

eral drugs with efficacy in migraine and no efficacy in any

other painful condition. This pertains to the 5-HT1B/D

receptor agonists, the triptans, CGRP receptor antagonists

and probably also for 5-HT1F receptor agonists [151, 152].

Because of their extreme receptor specificity these drugs

provide excellent validation of acute migraine models

[153]. Ergot alkaloids, ergotamine and dihydroergotamine,

are also specific for migraine [154, 155] but interact with a

multitude of receptors [155]. Thus, their mechanisms of

action are difficult to sort out and they are therefore of less

use. Non-steroidal anti-inflammatory drugs (NSAID) are

Table 4 continued

Effect observed Treatment Inhibition of effect

studied by treatment

References

GTN 80 lg/kg over

20 min iv

Awake rats

Increase in c-Fos expression in TNC

Increase in pERK in dura mater, TG, TNC

Increase CamKII in TNC

Increase in ATF1 in TNC

Increase in pCREB in TNC

[197]

GTN 10 lg/kg iv

over 20 min

Anesthetized

No effect on c-Fos expression in TNC [198]

GTN 10 mg/kg sc

(mice)

Increase in c-Fos expression in TNC [105]

Induces thermal allodynia Sumatriptan

Intrathecal 0.06 lg ?

ip 300 lg/kg –

ip 600 lg/kg ?

Induces mechanical allodynia Sumatriptan

Intrathecal 0.06 lg ?

ip 300 lg/kg –

ip 600 lg/kg –

No effect on CSD threshold

GTN 60 lg/kg iv

Anesthetized

Increase in IL1b expression in dura mater

Increase in iNOS expression in dura mater

Increase in mastcell degranulation

[113]

The experiment is performed in rats if species is not given

Significant effect ? P \ 0.05, ?? P \ 0.01, ??? P \ 0.001

1060 I. Jansen-Olesen et al.

Page 13: Animal Migraine Models for Drug Development: Status and Future Perspectives

effective in migraine [156] but also have efficacy in all

other painful conditions and are thus not suitable for vali-

dating a migraine model. A valid animal model should be

sensitive and specific. As with diagnostic criteria for dis-

eases too rigorous criteria result in too low sensitivity but

high specificity. As a best compromise we suggest that a

useful model should respond to at least two of the three

classes of specific anti-migraine drugs. Alternatively, one

might require that all specific drugs should work in the

model and no unspecific drugs should work. This would be

a very strict requirement and it seems unlikely that any

animal model would be able to deliver such results. It is

more likely that a model would respond to one or two of

the specific drugs but not to the others and that a number of

different models must be used to test future anti-migraine

drugs. Depending on the type of new drug one model might

respond and another not. The existence of a number of such

models would allow a relatively precise estimation of new

drugs for the acute treatment of migraine.

8 Validating Models of Prophylactic Migraine

Treatment

While models for the testing of acute migraine drugs are

available some of which have shown validity [27, 157], this

is not the case for models testing prophylactic anti-

migraine drugs. All available prophylactic compounds are

unspecific with multiple actions in the body and almost all

drugs have been developed for another indication and have

subsequently received migraine as a secondary indication

[6–8]. Furthermore, mechanisms of action of the prophy-

lactic anti-migraine drugs are highly variable and in gen-

eral not understood. Some antiepileptic drugs have for

example efficacy and others not. The same is true for anti-

hypersensitive drugs. Drugs that only have the migraine

indication such as flunarizine and pizotifen interact with

multiple receptor systems and their mode of action in

migraine is unknown [158, 159]. Adding to these problems

Fig. 5 Fos-positive cells in the superficial lamina (I/II) of rats treated

with saline, glyceryltrinitrate (GTN) and GTN in the presence of

sumatriptan (suma). Saline and one of the groups with GTN treated

rats were killed 2 h after the end of GTN infusion while the other

GTN treated group and the group with GTN treatment in the presence

of sumatriptan 0.6 mg/kg were killed at 4 h after termination of GTN

infusion. Six sections per animal evenly distributed from 0.8 to

5.12 mm distance from obex were counted for Fos-immunoreactivity.

Data are presented as mean ± SEM from 4–6 animals. Statistical

analysis using Kruskal–Wallis non parametric test **P \ 0.01 as

compared to saline treatment. Mann–Whitney U test was used

comparing the effect of sumatriptan on Fos activation induced by

GTN to GTN alone, #P \ 0.05 (adapted from Ramachandran et al.

[27])

Fig. 6 Sustained exposure to triptans reduces sensory thresholds to

light tactile stimuli applied to the periorbital region and the hind paws

of rats. Continuous infusion of sumatriptan (0.6 mg/kg/day sc)

decreased withdrawal thresholds to light tactile stimuli applied to

a the periorbital region or b the hind paws of rats. Sumatriptan or

saline was continuously administered through an osmotic minipump

for 6 days, after which the minipumps were removed. Withdrawal

responses to von Frey filaments were significantly (P \ 0.05) reduced

in a time dependent manner. BL baseline

Animal Migraine Models 1061

Page 14: Animal Migraine Models for Drug Development: Status and Future Perspectives

is the relatively low efficacy of prophylactic anti-migraine

drugs. Fifty percent efficacy in fifty percent of the patients

is the standard, with a therapeutic gain of 25 %. No drug

has been shown to be more effective than another [6, 7,

156, 158, 159]. Thus, it is difficult to use existing pro-

phylactic drugs for the validation of animal models. The

one exception may be CSD which is a valid model of

migraine with aura but not necessarily of migraine without

aura. CSD is strongly inhibited by tonabersat [65, 160]

which has efficacy in the prophylactic treatment of

migraine with aura [66] but not migraine without aura [67].

We suggest that a model for the development of prophy-

lactic drugs should be tested with propranolol and valpro-

ate. Propranolol is one of the oldest prophylactic migraine

drugs [8] and represents the class of antihypertensive

agents while valproate is the oldest within the group of

antiepileptic drugs [7]. Both drugs can be given as injec-

tion. In the human GTN model there was no preventive

efficacy of propranolol [161] but valproate was effective

[162]. Provocation with GTN may, however, affect

migraine mechanisms deep in the cascade of events while

propranolol may work higher in the cascade and valproate

deeper in the cascade. Such is currently only speculation

but the human findings illustrate that a battery of animal

models may be necessary.

9 The Future of Animal Migraine Models

The future of animal models for migraine looks relatively

bright. There are already several models responding to

triptan treatment and the cortical spreading depression

model mimics migraine with aura. There is no doubt that

further development is needed and these needs have been

discussed above. One problem is that migraine research is

much more poorly funded than research into any of the

other major neurological disorders [163]. While it is easy

to see what needs to be done, it will take considerable

resources to develop a combination of models that is

generally accepted as predictive of efficacy of anti-

migraine drugs. A major EU program or NIH program

would enhance such development considerably. Hopefully,

this paper has demonstrated that there is a lot of knowledge

about migraine mechanisms that can be used in the

development of more predictive models of migraine.

Among the major future lines of development, genetics

holds promise. The so-called familial hemiplegic migraine

mouse expressing a CACNA1A gene mutation causing

familial hemiplegic migraine (FHM) has already been used

in a number of elegant experiments [30, 34, 164]. Even if

this mouse model may not predict efficacy in migraine

without aura and migraine with typical aura, there might be

new models developed as more and more variants are

described that cause an increased risk of the prevalent types

of migraine. Thirteen variants have already been associated

with an increased risk of migraine Anttila et al. [199, 200]

but the increase in risk may not be high enough for a

knock-in model to be useful. The genetic variability of

migraine may also prove too large for useful animal models

of genetically modified mice. Cell lines expressing known

and future genetic variants looks like a more promising

way forward and the development of induced pluripotent

stem cells from migraine patients is actually part of a huge

program under the Innovative Medicines Initiative in

Europe called STEMBANCC.

We favour the use of stimulation of animals with agents

known to induce migraine in migraine patients and the use

of behavioural responses as outcome parameters. The fact

that provoking agents have proven ability to cause

migraine in migraine sufferers provides validity and

recording behavioural responses is associated with a rela-

tively high throughput allowing multiple doses to be tested

in the same animal in cross-over experiments. Maybe the

discovery of animals with particular characteristics will

revolutionize the whole field in the future. This may be

particularly true for the testing of prophylactic anti-

migraine drugs.

10 Conclusion

Many animal models of migraine have been proposed.

Most have, however, been used in the analysis of migraine

pathophysiology. Others have responded to single anti

migraine treatments but none have been thoroughly vali-

dated. A dedicated effort should be made to develop

models that respond to at least two existing anti migraine

drugs and not to drugs proposed for migraine but subse-

quently proved not to be effective.

Acknowledgments Inger Jansen-Olesen’s salary is paid by funding

from The Lundbeck Foundation and Candy’s Foundation. No other

funding has been received for this paper. Dr Inger Jansen-Olesen, Dr

Peer Tfelt-Hansen and Professor Jes Olesen declare that they have no

conflicts of interest.

References

1. Murray CJ, Vos T, Lozano R, Naghavi M, Flaxman AD, Mi-

chaud C, et al. Disability-adjusted life years (DALYs) for 291

diseases and injuries in 21 regions, 1990–2010: a systematic

analysis for the Global Burden of Disease Study 2010. Lancet.

2012;380(9859):2197–223.

2. Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati

M, et al. Years lived with disability (YLDs) for 1160 sequelae of

289 diseases and injuries 1990–2010: a systematic analysis for

the Global Burden of Disease Study 2010. Lancet.

2012;380(9859):2163–96.

1062 I. Jansen-Olesen et al.

Page 15: Animal Migraine Models for Drug Development: Status and Future Perspectives

3. Steiner T, Stovner L, Birbeck G. Migraine: the seventh disabler.

Cephalalgia. 2013;33(5):289–90.

4. Olesen J, Gustavsson A, Svensson M, Wittchen HU, Jonsson B.

The economic cost of brain disorders in Europe. Eur J Neurol.

2012;19(1):155–62.

5. Tfelt-Hansen P, Olesen J. Taking the negative view of migraine

treatments: the unmet needs. CNS Drugs. 2012;26(5):375–82.

6. Evers S, Mylecharane EJ. Nonsteroidal anti-inflammatory and

miscellaneous drugs in migraine prophylaxis. In: Olesen J,

Goadsby PJ, Ramadan NM, Tfelt-Hansen P, Welch KMA, edi-

tors. The headaches. 3 ed. Philadelphia: Lippincott, Williams &

Wilkins; 2006. p. 553–6.

7. Silberstein SD, Tfelt-Hansen P. Antiepileptic drugs in migraine

prophylaxis. In: Olesen J, Goadsby PJ, Ramadan NM, Tfelt-

Hansen P, Welch KMA, editors. The headaches. 3 ed. Phila-

delphia: Lippincott, Williams & Wilkins; 2006. p. 545–51.

8. Tfelt-Hansen P, Rolan P. beta-Adrenoceptor blocking drugs in

migraine prophylaxis. In: Olesen J, Goadsby PJ, Ramadan NM,

Tfelt-Hansen P, Welch KMA, editors. The headaches. 3 ed.

Philadelphia: Lippincott, Williams & Wilkins; 2006. p. 519–28.

9. Lipton RB, Bigal ME, Diamond M, Freitag F, Reed ML, Stewart

WF. Migraine prevalence, disease burden, and the need for

preventive therapy. Neurology. 2007;68(5):343–9.

10. Mergenthaler P, Meisel A. Do stroke models model stroke? Dis

Model Mech. 2012;5(6):718–25.

11. Cook DJ, Tymianski M. Translating promising preclinical

neuroprotective therapies to human stroke trials. Expert Rev

Cardiovasc Ther. 2011;9(4):433–49.

12. Gladstone DJ, Black SE, Hakim AM. Toward wisdom from

failure: lessons from neuroprotective stroke trials and new

therapeutic directions. Stroke. 2002;33(8):2123–36.

13. Andreou AP, Summ O, Charbit AR, Romero-Reyes M, Goadsby

PJ. Animal models of headache: from bedside to bench and back

to bedside. Expert Rev Neurother. 2010;10(3):389–411.

14. Bergerot A, Holland PR, Akerman S, Bartsch T, Ahn AH,

Maassenvandenbrink A, et al. Animal models of migraine:

looking at the component parts of a complex disorder. Eur J

Neurosci. 2006;24(6):1517–34.

15. Gupta S, Villalon CM. The relevance of preclinical research

models for the development of antimigraine drugs: focus on

5-HT(1B/1D) and CGRP receptors. Pharmacol Ther. 2010;

128(1):170–90.

16. Olesen J, Ashina M. Emerging migraine treatments and drug

targets. Trends Pharmacol Sci. 2011;32(6):352–9.

17. Ploug KB, Sorensen MA, Strobech L, Klaerke DA, Hay-

Schmidt A, Sheykhzade M, et al. K ATP channels in pig and

human intracranial arteries. Eur J Pharmacol. 2008;601(1–3):

43–9.

18. Myren M, Baun M, Ploug KB, Jansen-Olesen I, Olesen J, Gupta

S. Functional and molecular characterization of prostaglandin

E2 dilatory receptors in the rat craniovascular system in rele-

vance to migraine. Cephalalgia. 2010;30(9):1110–22.

19. Jansen-Olesen I, Kaarill L, Edvinsson L. Characterization of

CGRP(1) receptors in the guinea pig basilar artery. Eur J

Pharmacol. 2001;414(2–3):249–58.

20. Baun M, Hay-Schmidt A, Edvinsson L, Olesen J, Jansen-Olesen

I. Pharmacological characterization and expression of VIP and

PACAP receptors in isolated cranial arteries of the rat. Eur J

Pharmacol. 2011;670(1):186–94.

21. Wahl M, Lauritzen M, Schilling L. Change of cerebrovascular

reactivity after cortical spreading depression in cats and rats.

Brain Res. 1987;411(1):72–80.

22. Williamson DJ, Hargreaves RJ, Hill RG, Shepheard SL. Intra-

vital microscope studies on the effects of neurokinin agonists

and calcitonin gene-related peptide on dural vessel diameter in

the anaesthetized rat. Cephalalgia. 1997;17(4):518–24.

23. Levy D, Burstein R, Strassman AM. Calcitonin gene-related

peptide does not excite or sensitize meningeal nociceptors:

implications for the pathophysiology of migraine. Ann Neurol.

2005;58(5):698–705.

24. Hoskin KL, Kaube H, Goadsby PJ. Central activation of the

trigeminovascular pathway in the cat is inhibited by dihydro-

ergotamine. A c-Fos and electrophysiological study. Brain.

1996;119(Pt 1):249–56.

25. Knight YE, Bartsch T, Kaube H, Goadsby PJ. P/Q-type calcium-

channel blockade in the periaqueductal gray facilitates trigem-

inal nociception: a functional genetic link for migraine? J Neu-

rosci. 2002;22(5):RC213.

26. Koulchitsky S, Fischer MJ, De CR, Schlechtweg PM, Mes-

slinger K. Biphasic response to nitric oxide of spinal trigeminal

neurons with meningeal input in rat—possible implications for

the pathophysiology of headaches. J Neurophysiol. 2004;92(3):

1320–8.

27. Ramachandran R, Bhatt DK, Ploug KB, Olesen J, Jansen-Olesen

I, Hay-Schmidt A, et al. A naturalistic glyceryl trinitrate infusion

migraine model in the rat. Cephalalgia. 2012;32(1):73–84.

28. Tassorelli C, Joseph SA. NADPH-diaphorase activity and Fos

expression in brain nuclei following nitroglycerin administra-

tion. Brain Res. 1995;695(1):37–44.

29. Martino G, Perkins MN. Tactile-induced ultrasonic vocalization

in the rat: a novel assay to assess anti-migraine therapies in vivo.

Cephalalgia. 2008;28(7):723–33.

30. Langford DJ, Bailey AL, Chanda ML, Clarke SE, Drummond

TE, Echols S, et al. Coding of facial expressions of pain in the

laboratory mouse. Nat Methods. 2010;7(6):447–9.

31. De Felice M, Ossipov MH, Wang R, Lai J, Chichorro J, Meng I,

et al. Triptan-induced latent sensitization: a possible basis for

medication overuse headache. Ann Neurol. 2010;67(3):325–37.

32. Gupta S, Akerman S, van den Maagdenberg AM, Saxena PR,

Goadsby PJ, van den Brink AM. Intravital microscopy on a

closed cranial window in mice: a model to study trigemino-

vascular mechanisms involved in migraine. Cephalalgia.

2006;26(11):1294–303.

33. Tottene A, Pivotto F, Fellin T, Cesetti T, van den Maagdenberg

AM, Pietrobon D. Specific kinetic alterations of human CaV2.1

calcium channels produced by mutation S218L causing familial

hemiplegic migraine and delayed cerebral edema and coma after

minor head trauma. J Biol Chem. 2005;280(18):17678–86.

34. van den Maagdenberg AM, Pietrobon D, Pizzorusso T, Kaja S,

Broos LA, Cesetti T, et al. A Cacna1a knocking migraine mouse

model with increased susceptibility to cortical spreading

depression. Neuron. 2004;41(5):701–10.

35. Tottene A, Fellin T, Pagnutti S, Luvisetto S, Striessnig J,

Fletcher C, et al. Familial hemiplegic migraine mutations

increase Ca(2?) influx through single human CaV2.1 channels

and decrease maximal CaV2.1 current density in neurons. Proc

Natl Acad Sci USA. 2002;99(20):13284–9.

36. Mathew R, Andreou AP, Chami L, Bergerot A, van den Ma-

agdenberg AM, Ferrari MD, et al. Immunohistochemical char-

acterization of calcitonin gene-related peptide in the trigeminal

system of the familial hemiplegic migraine 1 knock-in mouse.

Cephalalgia. 2011;31(13):1368–80.

37. Hansen JM, Thomsen LL, Olesen J, Ashina M. Calcitonin gene-

related peptide does not cause the familial hemiplegic migraine

phenotype. Neurology. 2008;71(11):841–7.

38. Hansen JM, Thomsen LL, Olesen J, Ashina M. Familial hemi-

plegic migraine type 1 shows no hypersensitivity to nitric oxide.

Cephalalgia. 2008;28(5):496–505.

39. Hansen JM, Thomsen LL, Marconi R, Casari G, Olesen J,

Ashina M. Familial hemiplegic migraine type 2 does not share

hypersensitivity to nitric oxide with common types of migraine.

Cephalalgia. 2008;28(4):367–75.

Animal Migraine Models 1063

Page 16: Animal Migraine Models for Drug Development: Status and Future Perspectives

40. Fioretti B, Catacuzzeno L, Sforna L, Gerke-Duncan MB, van

den Maagdenberg AM, Franciolini F, et al. Trigeminal ganglion

neuron subtype-specific alterations of Ca(V)2.1 calcium current

and excitability in a Cacna1a mouse model of migraine.

J Physiol. 2011;589(Pt 23):5879–95.

41. Russo AF, Kuburas A, Kaiser EA, Raddant AC, Recober A. A

potential preclinical migraine model: CGRP-sensitized mice.

Mol Cell Pharmacol. 2009;1(5):264–70.

42. Recober A, Kuburas A, Zhang Z, Wemmie JA, Anderson MG,

Russo AF. Role of calcitonin gene-related peptide in light-

aversive behavior: implications for migraine. J Neurosci.

2009;29(27):8798–804.

43. Oshinsky ML, Sanghvi MM, Maxwell CR, Gonzalez D, Span-

genberg RJ, Cooper M, et al. Spontaneous trigeminal allodynia

in rats: a model of primary headache. Headache. 2012;52(9):

1336–49.

44. Markowitz S, Saito K, Moskowitz MA. Neurogenically medi-

ated plasma extravasation in dura mater: effect of ergot alka-

loids. A possible mechanism of action in vascular headache.

Cephalalgia. 1988;8(2):83–91.

45. Moskowitz MA. Neurogenic inflammation in the pathophysiol-

ogy and treatment of migraine. Neurology. 1993;43(6 Suppl

3):S16–20.

46. Markowitz S, Saito K, Moskowitz MA. Neurogenically medi-

ated leakage of plasma protein occurs from blood vessels in dura

mater but not brain. J Neurosci. 1987;7(12):4129–36.

47. Schuh-Hofer S, Siekmann W, Offenhauser N, Reuter U, Arnold

G. Effect of hyperoxia on neurogenic plasma protein extrava-

sation in the rat dura mater. Headache. 2006;46(10):1545–51.

48. Moskowitz MA, Macfarlane R. Neurovascular and molecular

mechanisms in migraine headaches. Cerebrovasc Brain Metab

Rev. 1993;5(3):159–77.

49. Lee WS, Limmroth V, Ayata C, Cutrer FM, Waeber C, Yu X,

et al. Peripheral GABAA receptor-mediated effects of sodium

valproate on dural plasma protein extravasation to substance P

and trigeminal stimulation. Br J Pharmacol. 1995;116(1):

1661–7.

50. Buzzi MG, Sakas DE, Moskowitz MA. Indomethacin and ace-

tylsalicylic acid block neurogenic plasma protein extravasation

in rat dura mater. Eur J Pharmacol. 1989;165(2–3):251–8.

51. Buzzi MG, Moskowitz MA. The antimigraine drug, sumatriptan

(GR43175), selectively blocks neurogenic plasma extravasation

from blood vessels in dura mater. Br J Pharmacol. 1990;99(1):202–6.

52. Brandli P, Loffler BM, Breu V, Osterwalder R, Maire JP, Clozel

M. Role of endothelin in mediating neurogenic plasma extrav-

asation in rat dura mater. Pain. 1996;64(2):315–22.

53. Connor HE, Bertin L, Gillies S, Beattie D, Ward P. Clinical

evaluation of a novel, potent, CNS penetrating NK1 receptor

antagonist in the acute treatment of migraine. Abstracts of the

12th migraine trust symposium, London; 1998.

54. Goldstein DJ, Wang O, Saper JR, Stoltz R, Silberstein SD,

Mathew NT. Ineffectiveness of neurokinin-1 antagonist in acute

migraine: a crossover study. Cephalalgia. 1997;17(7):785–90.

55. Goldstein DJ, Offen WW, Klein EG, Phebus LA, Hipskind P,

Johnson KW, et al. Lanepitant, an NK-1 antagonist, in migraine

prevention. Cephalalgia. 2001;21(2):102–6.

56. May A, Gijsman HJ, Wallnofer A, Jones R, Diener HC, Ferrari

MD. Endothelin antagonist bosentan blocks neurogenic inflam-

mation, but is not effective in aborting migraine attacks. Pain.

1996;67(2–3):375–8.

57. Norman B, Panebianco D, Block G. A placebo-controlled, in-

clinic study to explore the safety and efficacy of intravenous

L-758,298 (a pro-drug of the NK1-receptor antagonist

L-754,030) in the acute treatment of migraine. Abstracts of the

12th migraine trust symposium, London; 1998.

58. Shepheard SL, Williamson DJ, Hill RG, Hargreaves RJ. The

non-peptide neurokinin1 receptor antagonist, RP 67580, blocks

neurogenic plasma extravasation in the dura mater of rats. Br J

Pharmacol. 1993;108(1):11–2.

59. Shepheard SL, Williamson DJ, Williams J, Hill RG, Hargreaves

RJ. Comparison of the effects of sumatriptan and the NK1

antagonist CP-99,994 on plasma extravasation in Dura mater

and c-fos mRNA expression in trigeminal nucleus caudalis of

rats. Neuropharmacology. 1995;34(3):255–61.

60. Williamson DJ, Hargreaves RJ, Hill RG, Shepheard SL.

Sumatriptan inhibits neurogenic vasodilation of dural blood

vessels in the anaesthetized rat—intravital microscope studies.

Cephalalgia. 1997;17(4):525–31.

61. Williamson DJ, Hargreaves RJ. Neurogenic inflammation in the

context of migraine. Microsc Res Tech. 2001;53(3):167–78.

62. Petersen KA, Birk S, Doods H, Edvinsson L, Olesen J. Inhibi-

tory effect of BIBN4096BS on cephalic vasodilatation induced

by CGRP or transcranial electrical stimulation in the rat. Br J

Pharmacol. 2004;143(6):697–704.

63. Leao AA. Spreading depression of activity in the cerebral cor-

tex. J Neurophysiol. 1944;7:359–90.

64. Lauritzen M. Pathophysiology of the migraine aura. The

spreading depression theory. Brain. 1994;117(Pt 1):199–210.

65. Chan WN, Evans JM, Hadley MS, Herdon HJ, Jerman JC,

Parsons AA, et al. Identification of (–)-cis-6-acetyl-4S-(3-

chloro-4-fluoro-benzoylamino)-3,4-dihydro-2,2-dimethyl-2H-

benzo[b]pyran-3S-ol as a potential antimigraine agent. Bioorg

Med Chem Lett. 1999;9(2):285–90.

66. Hauge AW, Asghar MS, Schytz HW, Christensen K, Olesen J.

Effects of tonabersat on migraine with aura: a randomised,

double-blind, placebo-controlled crossover study. Lancet Neu-

rol. 2009;8(8):718–23.

67. Goadsby PJ, Ferrari MD, Csanyi A, Olesen J, Mills JG. Ran-

domized, double-blind, placebo-controlled, proof-of-concept

study of the cortical spreading depression inhibiting agent ton-

abersat in migraine prophylaxis. Cephalalgia. 2009;29(7):

742–50.

68. Parsons AA. Cortical spreading depression: its role in migraine

pathogenesis and possible therapeutic intervention strategies.

Curr Pain Headache Rep. 2004;8(5):410–6.

69. Afridi SK, Matharu MS, Lee L, Kaube H, Friston KJ, Frac-

kowiak RS, et al. A PET study exploring the laterality of

brainstem activation in migraine using glyceryl trinitrate. Brain.

2005;128(Pt 4):932–9.

70. Weiller C, May A, Limmroth V, Juptner M, Kaube H, Schayck

RV, et al. Brain stem activation in spontaneous human migraine

attacks. Nat Med. 1995;1(7):658–60.

71. Ayata C, Jin H, Kudo C, Dalkara T, Moskowitz MA. Suppres-

sion of cortical spreading depression in migraine prophylaxis.

Ann Neurol. 2006;59(4):652–61.

72. Bogdanov VB, Multon S, Chauvel V, Bogdanova OV, Prodanov

D, Makarchuk MY, et al. Migraine preventive drugs differen-

tially affect cortical spreading depression in rat. Neurobiol Dis.

2011;41(2):430–5.

73. Goadsby PJ, Zagami AS. Stimulation of the superior sagittal

sinus increases metabolic activity and blood flow in certain

regions of the brainstem and upper cervical spinal cord of the

cat. Brain. 1991;114(Pt 2):1001–11.

74. Goadsby PJ, Hoskin KL. Inhibition of trigeminal neurons by

intravenous administration of the serotonin (5HT)1B/D receptoragonist zolmitriptan (311C90): are brain stem sites therapeutic

target in migraine? Pain. 1996;67(2–3):355–9.

75. Hoskin KL, Goadsby PJ. Comparison of more and less lipophilic

serotonin (5HT1B/1D) agonists in a model of trigeminovascular

nociception in cat. Exp Neurol. 1998;150(1):45–51.

1064 I. Jansen-Olesen et al.

Page 17: Animal Migraine Models for Drug Development: Status and Future Perspectives

76. Kaube H, Hoskin KL, Goadsby PJ. Intravenous acetylsalicylic

acid inhibits central trigeminal neurons in the dorsal horn of the

upper cervical spinal cord in the cat. Headache. 1993;33(10):

541–4.

77. Shields KG, Goadsby PJ. Propranolol modulates trigeminovas-

cular responses in thalamic ventroposteromedial nucleus: a role

in migraine? Brain. 2005;128(Pt 1):86–97.

78. Storer RJ, Akerman S, Goadsby PJ. Calcitonin gene-related

peptide (CGRP) modulates nociceptive trigeminovascular

transmission in the cat. Br J Pharmacol. 2004;142(7):1171–81.

79. Storer RJ, Goadsby PJ. Topiramate inhibits trigeminovascular

neurons in the cat. Cephalalgia. 2004;24(12):1049–56.

80. Hoskin KL, Zagami AS, Goadsby PJ. Stimulation of the middle

meningeal artery leads to Fos expression in the trigeminocer-

vical nucleus: a comparative study of monkey and cat. J Anat.

1999;194(Pt 4):579–88.

81. Schepelmann K, Ebersberger A, Pawlak M, Oppmann M,

Messlinger K. Response properties of trigeminal brain stem

neurons with input from dura mater encephali in the rat. Neu-

roscience. 1999;90(2):543–54.

82. Goadsby PJ, Hoskin KL. Differential effects of low dose

CP122,288 and eletriptan on fos expression due to stimulation of

the superior sagittal sinus in cat. Pain. 1999;82(1):15–22.

83. Hoskin KL, Bulmer DC, Goadsby PJ. Fos expression in the

trigeminocervical complex of the cat after stimulation of the

superior sagittal sinus is reduced by L-NAME. Neurosci Lett.

1999;266(3):173–6.

84. Storer RJ, Akerman S, Shields KG, Goadsby PJ. GABAA

receptor modulation of trigeminovascular nociceptive neuro-

transmission by midazolam is antagonized by flumazenil. Brain

Res. 2004;1013(2):188–93.

85. Kaube H, Hoskin KL, Goadsby PJ. Inhibition by sumatriptan of

central trigeminal neurones only after blood–brain barrier dis-

ruption. Br J Pharmacol. 1993;109(3):788–92.

86. Goadsby PJ, Hoskin KL, Knight YE. Substance P blockade with

the potent and centrally acting antagonist GR205171 does not

effect central trigeminal activity with superior sagittal sinus

stimulation. Neuroscience. 1998;86(1):337–43.

87. Knight YE, Edvinsson L, Goadsby PJ. 4991W93 inhibits release

of calcitonin gene-related peptide in the cat but only at doses

with 5HT(1B/1D) receptor agonist activity? Neuropharmacol-

ogy. 2001;40(4):520–5.

88. Bartsch T, Goadsby PJ. Increased responses in trigeminocervical

nociceptive neurons to cervical input after stimulation of the

dura mater. Brain. 2003;126(8):1801–13.

89. Burstein R, Yamamura H, Malick A, Strassman AM. Chemical

stimulation of the intracranial dura induces enhanced responses

to facial stimulation in brain stem trigeminal neurons. J Neuro-

physiol. 1998;79(2):964–82.

90. Wieseler J, Ellis A, Sprunger D, Brown K, McFadden A, Ma-

honey J, et al. A novel method for modeling facial allodynia

associated with migraine in awake and freely moving rats.

J Neurosci Methods. 2010;185(2):236–45.

91. Yu XM, Sessle BJ, Vernon H, Hu JW. Effects of inflammatory

irritant application to the rat temporomandibular joint on jaw

and neck muscle activity. Pain. 1995;60(2):143–9.

92. Christiansen I, Thomsen LL, Daugaard D, Ulrich V, Olesen J.

Glyceryl trinitrate induces attacks of migraine without aura in

sufferers of migraine with aura. Cephalalgia. 1999;19(7):660–7.

93. Iversen HK, Olesen J, Tfelt-Hansen P. Intravenous nitroglycerin

as an experimental model of vascular headache. Basic charac-

teristics. Pain. 1989;38(1):17–24.

94. Schytz HW, Birk S, Wienecke T, Kruuse C, Olesen J, Ashina M.

PACAP38 induces migraine-like attacks in patients with

migraine without aura. Brain. 2009;132(Pt 1):16–25.

95. Schytz HW, Olesen J, Ashina M. The PACAP receptor: a novel

target for migraine treatment. Neurotherapeutics. 2010;7(2):

191–6.

96. Thomsen LL, Kruuse C, Iversen HK, Olesen J. A nitric oxide

donor (nitroglycerin) induces genuine migraine attacks. Eur J

Neurol. 1994;1:73–80.

97. Kruuse C, Thomsen LL, Jacobsen TB, Olesen J. The phospho-

diesterase 5 inhibitor sildenafil has no effect on cerebral blood

flow or blood velocity, but nevertheless induces headache in

healthy subjects. J Cereb Blood Flow Metab. 2002;22(9):

1124–31.

98. Kruuse C, Thomsen LL, Birk S, Olesen J. Migraine can be

induced by sildenafil without changes in middle cerebral artery

diameter. Brain. 2003;126(Pt 1):241–7.

99. Kruuse C, Lassen LH, Iversen HK, Oestergaard S, Olesen J.

Dipyridamole may induce migraine in patients with migraine

without aura. Cephalalgia. 2006;26(8):925–33.

100. Lassen LH, Thomsen LL, Olesen J. Histamine induces migraine

via the H1-receptor. Support for the NO hypothesis of migraine.

Neuroreport. 1995;6(11):1475–9.

101. Lassen LH, Haderslev PA, Jacobsen VB, Iversen HK, Sperling

B, Olesen J. CGRP may play a causative role in migraine.

Cephalalgia. 2002;22(1):54–61.

102. Krabbe AA, Olesen J. Headache provocation by continuous

intravenous infusion of histamine. Clinical results and receptor

mechanisms. Pain. 1980;8(2):253–9.

103. Meng ID, Bereiter DA. Differential distribution of Fos-like

immunoreactivity in the spinal trigeminal nucleus after noxious

and innocuous thermal and chemical stimulation of rat cornea.

Neuroscience. 1996;72(1):243–54.

104. Sugiyo S, Uehashi D, Satoh F, Abe T, Yonehara N, Kobayashi

M, et al. Effects of systemic bicuculline or morphine on for-

malin-evoked pain-related behaviour and c-Fos expression in

trigeminal nuclei after formalin injection into the lip or tongue

in rats. Exp Brain Res. 2009;196(2):229–37.

105. Bates E, Nikai T, Brennan K, Fu YH, Charles A, Basbaum A,

et al. Sumatriptan alleviates nitroglycerin-induced mechanical

and thermal allodynia in mice. Cephalalgia. 2010;30:170–8.

106. Knyihar-Csillik E, Vecsei L. Effect of a nitric oxide donor on

nitroxergic nerve fibers in the rat dura mater. Neurosci Lett.

1999;260(2):97–100.

107. Pardutz A, Krizbai I, Multon S, Vecsei L, Schoenen J. Systemic

nitroglycerin increases nNOS levels in rat trigeminal nucleus

caudalis. Neuroreport. 2000;11(14):3071–5.

108. Tassorelli C, Joseph SA, Nappi G. Central effects of nitro-

glycerin in the rat: new perspectives in migraine research. Funct

Neurol. 1996;11(5):219–35.

109. Tassorelli C, Joseph SA, Buzzi MG, Nappi G. The effects on the

central nervous system of nitroglycerin—putative mechanisms

and mediators. Prog Neurobiol. 1999;57(6):607–24.

110. Tassorelli C, Costa A, Blandini F, Joseph SA, Nappi G. Effect of

nitric oxide donors on the central nervous system—nitroglycerin

studies in the rat. Funct Neurol. 2000;15(Suppl 3):19–27.

111. Tassorelli C, Blandini F, Costa A, Preza E, Nappi G. Nitro-

glycerin-induced activation of monoaminergic transmission in

the rat. Cephalalgia. 2002;22(3):226–32.

112. Zhang X, Kainz V, Zhao J, Strassman AM, Levy D. Vascular

ERK mediates migraine-related sensitization of meningeal

nociceptors. Ann Neurol. 2013;73:741–50.

113. Reuter U, Bolay H, Jansen-Olesen I, Chiarugi A, del Sanchez

RM, Letourneau R, et al. Delayed inflammation in rat meninges:

implications for migraine pathophysiology. Brain. 2001;124(Pt

12):2490–502.

114. Read SJ, Smith MI, Hunter AJ, Parsons AA. Enhanced nitric

oxide release during cortical spreading depression following

Animal Migraine Models 1065

Page 18: Animal Migraine Models for Drug Development: Status and Future Perspectives

infusion of glyceryl trinitrate in the anaesthetized cat. Cepha-

lalgia. 1997;17(3):159–65.

115. Read SJ, Manning P, McNeil CJ, Hunter AJ, Parsons AA.

Effects of sumatriptan on nitric oxide and superoxide balance

during glyceryl trinitrate infusion in the rat. Implications for

antimigraine mechanisms. Brain Res. 1999;847(1):1–8.

116. Offenhauser N, Zinck T, Hoffmann J, Schiemann K, Schuh-

Hofer S, Rohde W, et al. CGRP release and c-fos expression

within trigeminal nucleus caudalis of the rat following glyce-

ryltrinitrate infusion. Cephalalgia. 2005;25(3):225–36.

117. Stenberg C, Ovlisen K, Svendsen O, Lauritzen B. Effect of local

anaesthesia on neuronal c-fos expression in the spinal dorsal

horn and hypothalamic paraventricular nucleus after surgery in

rats. Basic Clin Pharmacol Toxicol. 2005;96(5):381–6.

118. Sommers MG, Nguyen NK, Veening JG, Vissers KC, Ritskes-

Hoitinga M, van Egmond J. Suppression of noxious-induced

c-fos expression in the rat lumbar spinal cord by isoflurane alone

or combined with fentanyl. Anesth Analg. 2008;106(4):1303–8

(table).

119. Tassorelli C, Joseph SA. Systemic nitroglycerin induces Fos

immunoreactivity in brainstem and forebrain structures of the

rat. Brain Res. 1995;682(1–2):167–81.

120. Daugaard D, Thomsen LL, Iversen HK, Olesen J. Delayed

migraine-like headache in healthy volunteers after a combina-

tion of acetazolamide and glyceryl trinitrate. Cephalalgia.

2009;29(12):1294–300.

121. Oshinsky ML, Gomonchareonsiri S. Episodic dural stimulation

in awake rats: a model for recurrent headache. Headache.

2007;47(7):1026–36.

122. Strassman AM, Raymond SA, Burstein R. Sensitization of

meningeal sensory neurons and the origin of headaches. Nature.

1996;384(6609):560–4.

123. Levy D, Jakubowski M, Burstein R. Disruption of communi-

cation between peripheral and central trigeminovascular neurons

mediates the antimigraine action of 5HT 1B/1D receptor ago-

nists. Proc Natl Acad Sci USA. 2004;101(12):4274–9.

124. Andreou AP, Shields KG, Goadsby PJ. GABA and valproate

modulate trigeminovascular nociceptive transmission in the

thalamus. Neurobiol Dis. 2010;37(2):314–23.

125. Storer RJ, Akerman S, Goadsby PJ. GABA receptors modulate

trigeminovascular nociceptive neurotransmission in the trigem-

inocervical complex. Br J Pharmacol. 2001;134(4):896–904.

126. Burstein R, Jakubowski M, Garcia-Nicas E, Kainz V, Bajwa Z,

Hargreaves R, et al. Thalamic sensitization transforms localized

pain into widespread allodynia. Ann Neurol. 2010;68(1):81–91.

127. Summ O, Charbit AR, Andreou AP, Goadsby PJ. Modulation of

nociceptive transmission with calcitonin gene-related peptide

receptor antagonists in the thalamus. Brain. 2010;133(9):

2540–8.

128. Day HE, Kryskow EM, Nyhuis TJ, Herlihy L, Campeau S.

Conditioned fear inhibits c-fos mRNA expression in the central

extended amygdala. Brain Res. 2008;1229:137–46.

129. Dragunow M, Faull R. The use of c-fos as a metabolic marker in

neuronal pathway tracing. J Neurosci Methods. 1989;29(3):

261–5.

130. VanElzakker M, Fevurly RD, Breindel T, Spencer RL. Envi-

ronmental novelty is associated with a selective increase in Fos

expression in the output elements of the hippocampal formation

and the perirhinal cortex. Learn Mem. 2008;15(12):899–908.

131. Crown ED. The role of mitogen activated protein kinase sig-

naling in microglia and neurons in the initiation and mainte-

nance of chronic pain. Exp Neurol. 2012;234(2):330–9.

132. Ji RR, Gereau RW IV, Malcangio M, Strichartz GR. MAP

kinase and pain. Brain Res Rev. 2009;60(1):135.

133. Amin FM, Asghar MS, Hougaard A, Hansen AE, Larsen VA, de

Koning PJ, et al. Magnetic resonance angiography of

intracranial and extracranial arteries in patients with spontane-

ous migraine without aura: a cross-sectional study. Lancet

Neurol. 2013;12(5):454–61.

134. Goadsby PJ, Charbit AR, Andreou AP, Akerman S, Holland PR.

Neurobiology of migraine. Neuroscience. 2009;161(2):327–41.

135. Charles A. Vasodilation out of the picture as a cause of migraine

headache. Lancet Neurol. 2013;12(5):419–20.

136. Williamson DJ, Shepheard SL, Hill RG, Hargreaves RJ. The

novel anti-migraine agent rizatriptan inhibits neurogenic dural

vasodilation and extravasation. Eur J Pharmacol. 1997;328(1):

61–4.

137. Amrutkar DV, Ploug KB, Olesen J, Jansen-Olesen I. Role for

voltage gated calcium channels in calcitonin gene-related pep-

tide release in the rat trigeminovascular system. Neuroscience.

2011;172:510–7.

138. Amrutkar DV, Ploug KB, Hay-Schmidt A, Porreca F, Olesen J,

Jansen-Olesen I. mRNA expression of 5-hydroxytryptamine 1B,

1D, and 1F receptors and their role in controlling the release of

calcitonin gene-related peptide in the rat trigeminovascular

system. Pain. 2012;153(4):830–8.

139. Burstein R, Yarnitsky D, Goor-Aryeh I, Ransil BJ, Bajwa ZH.

An association between migraine and cutaneous allodynia. Ann

Neurol. 2000;47(5):614–24.

140. Kaube H, Katsarava Z, Przywara S, Drepper J, Ellrich J, Diener

HC. Acute migraine headache: possible sensitization of neurons

in the spinal trigeminal nucleus? Neurology. 2002;58(8):

1234–8.

141. Lipton RB, Bigal ME, Ashina S, Burstein R, Silberstein S, Reed

ML, et al. Cutaneous allodynia in the migraine population. Ann

Neurol. 2008;63(2):148–58.

142. Edelmayer RM, Ossipov MH, Porreca F. An experimental

model of headache-related pain. Methods Mol Biol. 2012;851:

109–20.

143. Zhang XC, Kainz V, Burstein R, Levy D. Tumor necrosis factor-

alpha induces sensitization of meningeal nociceptors mediated

via local COX and p38 MAP kinase actions. Pain. 2011;152(1):

140–9.

144. Strassman A, Mason P, Moskowitz M, Maciewicz R. Response

of brainstem trigeminal neurons to electrical stimulation of the

dura. Brain Res. 1986;379(2):242–50.

145. Thomsen LL, Brennum J, Iversen HK, Olesen J. Effect of a

nitric oxide donor (glyceryl trinitrate) on nociceptive thresholds

in man. Cephalalgia. 1996;16(3):169–74.

146. Markovics A, Kormos V, Gaszner B, Lashgarara A, Szoke E,

Sandor K, et al. Pituitary adenylate cyclase-activating poly-

peptide plays a key role in nitroglycerol-induced trigeminovas-

cular activation in mice. Neurobiol Dis. 2012;45(1):633–44.

147. De CD, Toldo I, Dal ZL, Perissinotto E, Sartori S, Gatta M, et al.

Osmophobia as an early marker of migraine: a follow-up study

in juvenile patients. Cephalalgia. 2012;32(5):401–6.

148. Zanchin G, Dainese F, Trucco M, Mainardi F, Mampreso E,

Maggioni F. Osmophobia in migraine and tension-type head-

ache and its clinical features in patients with migraine. Cepha-

lalgia. 2007;27(9):1061–8.

149. Kelman L. Osmophobia and taste abnormality in migraineurs: a

tertiary care study. Headache. 2004;44(10):1019–23.

150. Neususs K, Neumann B, Steinhoff BJ, Thegeder H, Bauer A,

Reimers D. Physical activity and fitness in patients with head-

ache disorders. Int J Sports Med. 1997;18(8):607–11.

151. Skingle M, Birch PJ, Leighton GE, Humphrey PP. Lack of an-

tinociceptive activity of sumatriptan in rodents. Cephalalgia.

1990;10(5):207–12.

152. Brennum J, Brinck T, Schriver L, Wanscher B, Sorensen PS,

Tfel-Hansen P, et al. Sumatriptan has no clinically relevant

effect in the treatment of episodic tensionCCEtype headache.

Eur J Neurol. 1996;3(1):23–8.

1066 I. Jansen-Olesen et al.

Page 19: Animal Migraine Models for Drug Development: Status and Future Perspectives

153. Hirsch S, Just S, Corradini L, Doods H. The CGRP receptor

antagonist BIBN4096BS peripherally alleviates inflammatory

pain in rats. Pain. 2013;154:700–7.

154. Tfelt-Hansen P. Ergotamine, dihydroergotamine: current uses

and problems. Curr Med Res Opin. 2001;17(Suppl 1):s30–4.

155. Tfelt-Hansen P, Saxena PR. Ergot alkaloids in the acute treat-

ment of migraine. In: Olesen J, Goadsby PJ, Ramadan NM,

Tfelt-Hansen P, Welch KMA, editors. The headaches. 3 ed.

Philadelphia: Lippincott, Williams & Wilkins; 2006. p. 459–67.

156. Tfelt-Hansen P, Rolan P. Nonsteroidal antiinflammatory drugs

in the acute treatment of migraine. In: Olesen J, Goadsby PJ,

Ramadan NM, Tfelt-Hansen P, Welch KMA, editors. The

headaches. 3 ed. Philadelphia: Lippincott, Williams & Wilkins;

2006. p. 449–57.

157. Ramachandran R, Bhatt DK, Ploug KB, Hay-Schmidt A, Jan-

sen-Olesen I, Gupta S, et al. Nitric oxide synthase, calcitonin

gene-related peptide and neurokinins are involved in GTN

induced neuronal activation. Cephalalgia. 2013 (in press).

158. Toda N, Tfelt-Hansen P. Calcium antagonists in migraine pro-

phylaxis. In: Olesen J, Goadsby PJ, Ramadan NM, Tfelt-Hansen

P, Welch KMA, editors. The headaches. 3 ed. Philadelphia:

Lippincott, Williams & Wilkins; 2006. p. 539–44.

159. Tfelt-Hansen P, Saxena PR. Antiserotonin drugs in migraine

prophylaxis. In: Olesen J, Goadsby PJ, Ramadan NM, Tfelt-

Hansen P, Welch KMA, editors. The headaches. 3 ed. Phila-

delphia: Lippincott, Williams & Wilkins; 2006. p. 529–37.

160. Read SJ, Hirst WD, Upton N, Parsons AA. Cortical spreading

depression produces increased cGMP levels in cortex and brain

stem that is inhibited by tonabersat (SB-220453) but not

sumatriptan. Brain Res. 2001;891(1–2):69–77.

161. Tvedskov JF, Thomsen LL, Thomsen LL, Iversen HK, Williams

P, Gibson A, et al. The effect of propranolol on glyceryltrini-

trate-induced headache and arterial response. Cephalalgia.

2004;24(12):1076–87.

162. Tvedskov JF, Thomsen LL, Iversen HK, Gibson A, Wiliams P,

Olesen J. The prophylactic effect of valproate on glyceryltrini-

trate induced migraine. Cephalalgia. 2004;24(7):576–85.

163. Sobocki P, Lekander I, Berwick S, Olesen J, Jonsson B.

Resource allocation to brain research in Europe (RABRE). Eur J

Neurosci. 2006;24(10):2691–3.

164. Chanda ML, Tuttle AH, Baran I, Atlin C, Guindi D, Hathaway

G, et al. Behavioral evidence for photophobia and stress-related

ipsilateral head pain in transgenic Cacna1a mutant mice. Pain.

2013;154:1254–62.

165. Matsubara T, Moskowitz MA, Huang Z. UK-14,304, R(–)-

alpha-methyl-histamine and SMS 201-995 block plasma protein

leakage within dura mater by prejunctional mechanisms. Eur J

Pharmacol. 1992;224(2–3):145–50.

166. O’Shaughnessy CT, Connor HE. Investigation of the role of

tachykinin NK1, NK2 receptors and CGRP receptors in neuro-

genic plasma protein extravasation in dura mater. Eur J Phar-

macol. 1994;263(1–2):193–8.

167. Shepherd SL, Williamson DJ, Beer MS, Hill RG, Hargreaves

RJ. Differential effects of 5-HT1B/1D receptor agonists on

neurogenic dural plasma extravasation and vasodilation in

anaesthetized rats. Neuropharmacology. 1997;36(4–5):525–33.

168. Cutrer FM, Yu XJ, Ayata G, Moskowitz MA, Waeber C. Effects

of PNU-109,291, a selective 5-HT1D receptor agonist, on

electrically induced dural plasma extravasation and capsaicin-

evoked c-fos immunoreactivity within trigeminal nucleus cau-

dalis. Neuropharmacology. 1999;38(7):1043–53.

169. Gozalov A, Jansen-Olesen I, Klaerke D, Olesen J. Role of

BK(Ca) channels in cephalic vasodilation induced by CGRP,

NO and transcranial electrical stimulation in the rat. Cephalal-

gia. 2007;27(10):1120–7.

170. Juhl L, Edvinsson L, Olesen J, Jansen-Olesen I. Effect of two

novel CGRP-binding compounds in a closed cranial window rat

model. Eur J Pharmacol. 2007;567(1–2):117–24.

171. Gozalov A, Jansen-Olesen I, Klaerke D, Olesen J. Role of K

ATP channels in cephalic vasodilatation induced by calcitonin

gene-related peptide, nitric oxide, and transcranial electrical

stimulation in the rat. Headache. 2008;48(8):1202–13.

172. Chan KY, Gupta S, de Vries R, Danser AH, Villalon CM,

Munoz-Islas E, et al. Effects of ionotropic glutamate receptor

antagonists on rat dural artery diameter in an intravital micros-

copy model. Br J Pharmacol. 2010;160(6):1316–25.

173. Bartsch T, Akerman S, Goadsby PJ. The ORL-1 (NOP1)

receptor ligand nociceptin/orphanin FQ (N/OFQ) inhibits neu-

rogenic dural vasodilatation in the rat. Neuropharmacology.

2002;43(6):991–8.

174. Akerman S, Williamson DJ, Kaube H, Goadsby PJ. Nitric oxide

synthase inhibitors can antagonize neurogenic and calcitonin

gene-related peptide induced dilation of dural meningeal ves-

sels. Br J Pharmacol. 2002;137(1):62–8.

175. Akerman S, Williamson DJ, Kaube H, Goadsby PJ. The effect of

anti-migraine compounds on nitric oxide-induced dilation of

dural meningeal vessels. Eur J Pharmacol. 2002;452(2):223–8.

176. Akerman S, Williamson DJ, Hill RG, Goadsby PJ. The effect of

adrenergic compounds on neurogenic dural vasodilatation. Eur J

Pharmacol. 2001;424(1):53–8.

177. Akerman S, Williamson DJ, Kaube H, Goadsby PJ. The role of

histamine in dural vessel dilation. Brain Res. 2002;956(1):

96–102.

178. Akerman S, Williamson DJ, Goadsby PJ. Voltage-dependent

calcium channels are involved in neurogenic dural vasodilata-

tion via a presynaptic transmitter release mechanism. Br J

Pharmacol. 2003;140(3):558–66.

179. Shepheard S, Edvinsson L, Cumberbatch M, Williamson D,

Mason G, Webb J, et al. Possible antimigraine mechanisms of

action of the 5HT1F receptor agonist LY334370. Cephalalgia.

1999;19(10):851–8.

180. Williamson DJ, Shepheard SL, Cook DA, Hargreaves RJ, Hill

RG, Cumberbatch MJ. Role of opioid receptors in neurogenic

dural vasodilation and sensitization of trigeminal neurones in

anaesthetized rats. Br J Pharmacol. 2001;133(6):807–14.

181. Holland PR, Akerman S, Goadsby PJ. Orexin 1 receptor acti-

vation attenuates neurogenic dural vasodilation in an animal

model of trigeminovascular nociception. J Pharmacol Exp Ther.

2005;315(3):1380–5.

182. Akerman S, Kaube H, Goadsby PJ. Anandamide is able to

inhibit trigeminal neurons using an in vivo model of trigemi-

novascular-mediated nociception. J Pharmacol Exp Ther.

2004;309(1):56–63.

183. Akerman S, Goadsby PJ. Topiramate inhibits trigeminovascular

activation: an intravital microscopy study. Br J Pharmacol.

2005;146(1):7–14.

184. Honey AC, Bland-Ward PA, Connor HE, Feniuk W, Humphrey

PP. Study of an adenosine A1 receptor agonist on trigeminally

evoked dural blood vessel dilation in the anaesthetized rat.

Cephalalgia. 2002;22(4):260–4.

185. Kaube H, Keay KA, Hoskin KL, Bandler R, Goadsby PJ.

Expression of c-Fos-like immunoreactivity in the caudal

medulla and upper cervical spinal cord following stimulation of

the superior sagittal sinus in the cat. Brain Res. 1993;629(1):

95–102.

186. Zagami AS, Goadsby PJ, Edvinsson L. Stimulation of the

superior sagittal sinus in the cat causes release of vasoactive

peptides. Neuropeptides. 1990;16(2):69–75.

187. Knight YE, Edvinsson L, Goadsby PJ. Blockade of calcitonin

gene-related peptide release after superior sagittal sinus

Animal Migraine Models 1067

Page 20: Animal Migraine Models for Drug Development: Status and Future Perspectives

stimulation in cat: a comparison of avitriptan and CP122,288.

Neuropeptides. 1999;33(1):41–6.

188. Benjamin L, Levy MJ, Lasalandra MP, Knight YE, Akerman S,

Classey JD, et al. Hypothalamic activation after stimulation of

the superior sagittal sinus in the cat: a Fos study. Neurobiol Dis.

2004;16(3):500–5.

189. Pardutz A, Multon S, Malgrange B, Parducz A, Vecsei L,

Schoenen J. Effect of systemic nitroglycerin on CGRP and 5-HT

afferents to rat caudal spinal trigeminal nucleus and its modu-

lation by estrogen. Eur J Neurosci. 2002;15(11):1803–9.

190. Pardutz A, Hoyk Z, Varga H, Vecsei L, Schoenen J. Oestrogen-

modulated increase of calmodulin-dependent protein kinase II

(CamKII) in rat spinal trigeminal nucleus after systemic nitro-

glycerin. Cephalalgia. 2007;27(1):46–53.

191. Knyihar-Csillik E, Toldi J, Krisztin-Peva B, Chadaide Z, Ne-

meth H, Fenyo R, et al. Prevention of electrical stimulation-

induced increase of c-fos immunoreaction in the caudal tri-

geminal nucleus by kynurenine combined with probenecid.

Neurosci Lett. 2007;418(2):122–6.

192. Knyihar-Csillik E, Mihaly A, Krisztin-Peva B, Robotka H,

Szatmari I, Fulop F, et al. The kynurenate analog SZR-72 pre-

vents the nitroglycerol-induced increase of c-fos immunoreac-

tivity in the rat caudal trigeminal nucleus: comparative studies

of the effects of SZR-72 and kynurenic acid. Neurosci Res.

2008;61(4):429–32.

193. Tassorelli C, Blandini F, Greco R, Nappi G. Nitroglycerin

enhances cGMP expression in specific neuronal and cerebro-

vascular structures of the rat brain. J Chem Neuroanat.

2004;27(1):23–32.

194. Tassorelli C, Greco R, Morazzoni P, Riva A, Sandrini G, Nappi

G. Parthenolide is the component of tanacetum parthenium that

inhibits nitroglycerin-induced Fos activation: studies in an ani-

mal model of migraine. Cephalalgia. 2005;25(8):612–21.

195. Greco R, Gasperi V, Sandrini G, Bagetta G, Nappi G, Mac-

carrone M, et al. Alterations of the endocannabinoid system in

an animal model of migraine: evaluation in cerebral areas of rat.

Cephalalgia. 2010;30(3):296–302.

196. Varga H, Pardutz A, Vamos E, Bohar Z, Bago F, Tajti J, et al.

Selective inhibition of cyclooxygenase-2 attenuates nitroglyc-

erin-induced calmodulin-dependent protein kinase II alpha in rat

trigeminal nucleus caudalis. Neurosci Lett. 2009;451(2):170–3.

197. Ramachandran R, Pedersen SH, Gupta S, Hay-Schmidt A,

Jansen-Olesen I, Olesen J. Downstream signalling mechanisms

in the trigeminovascular system of awake rats after glyceryl

trinitrate (GTN) infusion. Brain Res. 2013 (submitted).

198. Martin RS, Martin GR. Investigations into migraine pathogen-

esis: time course for effects of m-CPP, BW723C86 or glyceryl

trinitrate on appearance of Fos-like immunoreactivity in rat

trigeminal nucleus caudalis (TNC). Cephalalgia. 2001;21(1):

46–52.

199. Anttila V, et al. Genome-wide association study of migraine

implicates a common susceptibility variant on 8q22.1. Nat

Genet. 2010;42:869–73

200. Anttila V, et al. Genome-wide meta-analysis identifies new

susceptibility loci for migraine. Nat Genet. 2013;45:912–7

1068 I. Jansen-Olesen et al.