Chronic Migraine Lab Method

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Current Pain and Headache Reports (2018) 22:44

https://doi.org/10.1007/s11916-018-0693-5

CHRONIC DAILY HEADACHE (SJ WANG, SECTION EDITOR)

Animal Models of Chronic Migraine


Tse-Ming Chou 1,2 & Shih-Pin Chen 1,3,4,5,6

# Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
Purpose of Review Chronic migraine (CM) is a recalcitrant subtype of migraine which causes high degrees of disability, poor
treatment responses, and frequent recurrences in sufferers. However, the pathophysiological mechanisms underlying the devel-
opment and chronification of migraine attacks remain incompletely understood. A validated animal model could help to decipher
the pathogenic mechanism of the disease, facilitating the development of possible therapeutic strategies for CM. In this review,
we aimed to summarize current animal models of CM and discuss the validity of these models.
Recent Findings Several methods have been available to induce recurrent headache-like behaviors or biochemical changes in
rodents, including repeated dural application of inflammatory soup, chronic systemic infusion of nitroglycerin, repeated admin-
istration of acute migraine abortive treatment to simulate medication overuse headache, or genetic modification. These models
exhibit some features that are believed to be associated with migraine; however, none of the model can recapitulate all the clinical
phenotypes found in humans and each has its own weakness.
Summary The complex features of CM increase the difficulty of constructing a proper animal model. Nonetheless, currently
available models are valid to certain degrees. Future directions might consider simulating the spontaneity and chronicity of
migraine by combining known genetic substrates and allostatic loads into the same model.

Keywords Cortical spreading depression . Chronic migraine . Medication overuse headache . Sensitization . Trigeminovascular
system

Introduction edition, (ICHD-3) defined CM as headache at a frequency of


≥ 15 days per month for ≥ 3 months, of which ≥ 8 attacks are
Chronic migraine (CM) is a highly disabling subtype of mi- migraine or are responsive to migraine-specific treatment [3].
graine, with a global prevalence of around 2–4% [1, 2]. The CM usually develops from episodic migraine, with a conver-
International Classification of Headache Disorders, 3rd sion rate of about 3% a year [4, 5••]. The pathophysiology of
migraine chronification is elusive. Some possible pathophys-
This article is part of the Topical Collection on Chronic Daily Headache iological mechanisms of CM have been proposed, including
(1) dysfunction of the descending pain-modulating network
* Shih-Pin Chen [6, 7], (2) altered trigeminal and cranial autonomic system
chensp1977@gmail.com
function [8–10], (3) thalamic contribution to central sensitiza-
1 tion [11–13], and (4) medication-associated central sensitiza-
Institute of Neuroscience, National Yang-Ming University,
Taipei, Taiwan tion [14–18]. Along with the sensitization of the
2 trigeminovascular system, some nuclei in pain modulation
Interdisciplinary Neuroscience Program, Taiwan International
Graduate Program, Academia Sinica, Taipei, Taiwan circuits, especially those in the brainstem (e.g., periaqueductal
3 gray, locus coeruleus, raphe nucleus, and rostroventral medul-
Institute of Clinical Medicine, National Yang-Ming University,
Taipei, Taiwan la) and diencephalon (including the hypothalamus and thala-
4 mus), might be sensitized [6, 19–23]. Nevertheless, the exact
Division of Translational Research, Department of Medical
Research, Taipei Veterans General Hospital, Taipei, Taiwan pathophysiological mechanisms of CM remain to be clarified.
5 Animal models have been employed widely to study the
Department of Neurology, Neurological Institute, Taipei Veterans
General Hospital, Taipei, Taiwan pathogeneses of multiple medical disorders. Experimental
6 models of migraine have been developed and explored in past
Brain Research Center, National Yang-Ming University,
Taipei, Taiwan decades, leading to significant advances in the understanding
44 Page 2 of 8 Curr Pain Headache Rep (2018) 22:44

of the potential mechanisms of the disorder and facilitating the comorbidities, such as anxiety and depression, which are
development of antimigraine treatments [24–29]. Although much worse in patients with CM [47–50]. Comorbidity with
none of these experimental animal models can reflect all of depression and anxiety may increase headache frequency in
the features of migraine, discoveries made with them have migraineurs [50–53]. In addition, a substantial proportion of
been successfully translated into migraine therapeutics, such patients with CM is comorbid with medication overuse head-
as triptans [30–33] and drugs targeting the calcitonin gene- ache (MOH) [54–56]. Thus, the phenotypes of a valid CM
related peptide (CGRP) and its receptor [34–36] (e.g., model should mimic at least some of these clinical features
CGRP receptor antagonists [37–39] and monoclonal antibod- and comorbidities. In recent preclinical studies, various ap-
ies against CGRP and its receptor [40–45••]). Thus, the con- proaches have achieved the portrayal of clinical traits similar
struction of animal models addressing distinct aspects of the to those of migraine, including cutaneous allodynia and me-
possible mechanisms of CM not only could advance our un- chanical hyperalgesia [57–62], photophobia [63], MOH
derstanding of the disease, but might also contribute to the [14–16, 18], and depression and anxiety [64]. However, the
development of therapeutic approaches to tackle this recalci- demonstration of all of these sub-phenotypes with a single
trant subtype of migraine. However, many fewer models have experimental paradigm is difficult. In addition, concern exists
specifically targeted CM compared with migraine in general. about whether animals can experience “spontaneous”
In this review, we aim to summarize the currently available migraine-like episodes as patients do, although behavioral
animal models of CM; analyze their validity, advantages, and studies in mice carrying the human familial hemiplegic mi-
disadvantages from various perspectives; and propose direc- graine type 1 (FHM1) mutation have produced evidence sug-
tions for further research. gestive of spontaneous head pain upon subjection to novelty
or restraint stress [65]. In one study, a subset of a colony of
Sprague-Dawley rats also showed spontaneous episodic tri-
Key Determinants for the Construction of an geminal allodynia [59]. Although far from ideal, the currently
Effective Animal Model for CM available animal models of CM have at least in part mimicked
migraine phenomenology. Nevertheless, much remains to be
A valid animal model acting as a surrogate for human disease done to refine these models or create new models using novel
should have five features: similarity of symptoms, observabil- neuroscience or bioengineering techniques.
ity and measurability of behavioral phenotypes, inter-observer
reliability of assessment results, similarity of response to treat- Representation of the Chronicity of the Disorder
ment, and reproducibility of the system [46]. Thus, an ideal
animal model of CM should reasonably mimic clinical fea- Pain chronicity is associated with peripheral reorganization of
tures in patients, exhibit the chronicity of the disorder, respond afferent signaling and changes in the sensitivity of nociceptors
to the pharmacological interventions that are known to be and, perhaps, tactile afferents [66]. Chronification of the dis-
effective for CM, and involve mechanisms similar to those ease is accompanied by sensitization of the central nervous
identified in patients. system, causing further cortical reorganization in the brain
Because of the complexity of migraine and the intrinsic [66, 67]. A similar pathogenesis has been proposed for CM
differences between animals and humans, fulfillment of all [68–70]. The activation and sensitization of the
of these criteria is difficult. The chronification and transfor- trigeminovascular system are considered to play central roles
mation of migraine are particularly enigmatic, despite the in the pathogenesis of individual migraine attacks. Activation
characterization of risk factors. Furthermore, whether animals of the trigeminal nociceptive nerve endings innervating the
can actually experience “migraine” is another concern. These cranial vasculature could lead to the release of vasoactive neu-
obstacles exemplify some of the challenges in constructing an ropeptides, such as substance P and CGRP, which results in
animal model for CM, which must be overcome to facilitate neurogenic inflammation at the cortico-meningeal level and
the development of novel diagnostic and therapeutic ap- subsequent sensitization of the trigeminal nociceptors [71].
proaches for CM. The following are the key determinants that Sensitized trigeminal nociceptive nerve endings increase input
we consider to be critical for the establishment of an effective to the trigeminal nucleus caudalis (TNC) via the trigeminal
animal model of CM. ganglion (TG), causing activation of the brain regions in-
volved in pain processing. The superior salivatory nucleus is
Recapitulation of the Clinical Features of Migraine also activated when the TNC is sensitized, leading to the di-
latation of meningeal arteries via parasympathetic innervation,
Migraine is a complex neurological disorder characterized by which further augments perivascular trigeminal nociceptive
typical throbbing and unilateral headache, accompanied by inputs [72–76]. In CM, the activation and sensitization of
nausea, vomiting, photophobia, and phonophobia [3]. the trigeminovascular system and related pain circuits recur
Patients with migraine are more likely to have psychiatric frequently and become persistent with disease chronification,
Curr Pain Headache Rep (2018) 22:44 Page 3 of 8 44

leading to the structural and functional reorganization of pain- [14–16, 18, 87] (Table 1). These models exhibit some features that
related circuits. Evidence for the brain signatures of are believed to be associated (strongly) with migraine, including
chronification remains scarce in currently available models. periorbital, perimasseter, or hindpaw mechanical hyperalgesia [58,
In addition, the criteria for CM, especially headache frequency 62, 64, 82, 83]; photophobia [63, 88]; reduced locomotor activity
(i.e., ≥ 15 headache days per month, of which ≥ 8 are migraine [10, 63, 82, 88]; nociceptive behaviors [10]; unilateral hindpaw
attacks) and duration (≥ 3 months), are defined arbitrarily. It facial grooming [10]; facial expressions of pain [63, 88]; anxiety-
remains to be explored which attack frequency and duration and depression-like behaviors [64]; bioelectrical alterations related
are sufficient for CM modeling, and how long “headache sur- to cortical spreading depression (CSD) [16, 18, 87, 89]; activation
rogates” should last when exogenously administered stimuli of inflammatory markers in the TG [14, 15, 87], increased Fos [16,
(e.g., inflammatory soup, nitroglycerin, or pain medicine) are 18, 90] or extracellular glutamate expression [62] in the TNC; and
discontinued. To reproduce the chronicity of the disorder in alteration of CGRP-related genes in the TG and TNC [82]
future models, all of these factors should be taken into (Table 1). These models are considered to be valid to certain
consideration. degrees. For example, a mouse model of chronic nitroglycerin-
induced hyperalgesia was responsive to the preventive migraine
Reproduction of the Effects of Pharmacological therapies topiramate [84•] and propranolol [58], fulfilling the “pre-
Interventions dictive validity” requirement of an eligible model. However, the
same model failed to demonstrate the effectiveness of valproic
As migraine progresses toward a chronic state, it becomes acid, another migraine preventive drug [58]. Rats receiving epidu-
more refractory to acute and chronic pharmacological treat- ral inflammatory soup stimulation exhibited a cutaneous
ments. This relative refractoriness to treatment is a great allodynia-like response and hyperresponsiveness to nitroglycerin
challenge in the modeling of CM. In addition, the lack of [62] as well as increased nociceptive behaviors and unilateral
knowledge about the pathophysiological mechanism and the hindpaw facial grooming [10], fulfilling the “face validity” (i.e.,
generalized nature of functional and structural brain abnor- analogy of symptoms) of the model, although whether this model
malities pose challenges in the search for potential treatments. could also fulfill construct validity (i.e., comparable etiology) is not
Currently, the only indicated acute migraine-specific therapies without doubt.
are triptans and ergots, and the efficacy of both is limited for The major critiques of these models are that they are not
CM. In fact, the effect of sumatriptan was found to differ naturally occurring and that the stimuli causing head pain (if
across models. In one mouse model in which chronic inter- any) are supraphysiological. For example, one might argue
mittent nitroglycerin administration was used as a migraine that the consequence of the repetitive epidural application of
trigger, sumatriptan failed to abolish mechanical hyperalgesia inflammatory soup is likely chemical pachymeningitis, in-
[58]. In a rat model involving similar intermittent nitroglycer- stead of migraine; the behavioral changes observed after re-
in administration, sumatriptan attenuated hypoactivity and re- peated administration of high-dose nitroglycerin might not be
duced facial expressions of pain [63]. Overuse of these acute due solely to its nitric oxide donor effect, but could also be
treatments could contribute to the chronification of migraine, attributable to its vehicles [88]; and MOH models might not
which might be taken as an advantage in the construction of a be equivalent to CM models, as migraine rarely develops after
CM model [14, 15, 61, 77], but could also complicate the medication overuse in patients with no pre-existing headache
system by introducing an effect that not all patients will expe- (although we cannot know for sure whether drug-naïve wild-
rience. Thus, a reliable model of CM should be predictively type rodents have pre-existing headache). In addition, respon-
validated with pharmacodynamic treatments that have been siveness to triptans is not consistent across models. In a mouse
proven to be effective in patients [26, 27], such as topiramate model of chronic intermittent nitroglycerin-evoked
[78] and the newly developed CGRP-targeting therapies hyperalgesia, sumatriptan was ineffective in inhibiting me-
[45••], serving as positive controls [79–81]. chanical hyperalgesia, but instead aggravated the headache
surrogate hindpaw hyperalgesia [58], similar to the triptan-
induced latent sensitization MOH model in rats [14].
Current Animal Models of CM However, sumatriptan inhibited hypoactivity and grimace
scale scores evoked by chronic nitroglycerin infusion in rats
Currently, several methods are available to induce recurrent [63]. In addition, zolmitriptan was found to effectively inhibit
headache-like behaviors or biochemical changes in experimental headache-like behaviors in rats treated with epidural inflam-
animals, including repeated dural stimulation via epidural applica- matory soup [10]. Chronic administration of sumatriptan [16,
tion of inflammatory soup [10, 61, 62, 64, 82, 83], chronic sys- 89] or paracetamol [18, 87] increased susceptibility to CSD in
temic infusion of the nitric oxide donor nitroglycerin [58, 63, 84•, rats, suggesting the development of cortical hyperexcitability
85, 86], and repeated administration of acute migraine abortive in these MOH models. However, chronic daily CSD (without
treatment (e.g., sumatriptan or paracetamol) to simulate MOH repeated use of acute medication) did not increase, but
44 Page 4 of 8 Curr Pain Headache Rep (2018) 22:44

Table 1 Currently available models of chronic migraine

Models Animals Route and Endpoints Response to migraine treatment Validity Shortcomings Reference
duration of no.
administration

Repeated Rat Epiural Mechanical Zolmitriptan reduced nociceptive Face Supraphysiological [10, 61, 62,
inflamma- cannulation; hyperalgesia behaviors validi- inflammation 64, 82,
tory soup 4, 7, ≧ 21, Reduced Ketorolac reduced the nociceptive ty: +++ Allodynia is not equivalent 83]
30 days locomotor behavior and ipsilateral hindpaw Predictive to spontaneous head pain
activities facial grooming validi-
Nociceptive Amitriptyline reversed the allodynia ty: ++
behavior and decreased depression- and Construct
Unilateral anxiety-like behaviors validi-
hindpaw facial ty: ?
grooming
Anxiety- and
depression-like
behaviors
Increased
extracellular
glutamate
expression in
the TNC
Altered
CGRP-related
genes in the TG
and TNC
Nitroglycerin Rat, Intraperitoneal; Mechanical Propranolol, topiramate, and amiloride Face Effect of vehicle [58, 63,
mouse 11, hyperalgesia inhibited mechanical hyperalgesia; validi- Possibility of hypotension 84••, 85,
> 14 days Photophobia valproic acid and memantine were ty: ++ Inconsistent response to 86]
Reduced ineffective Predictive migraine treatment
locomotor Sumatriptan inhibited hypoactivity and validi- across models
activities grimace scale scores in rats but ty: +/− Allodynia is not equivalent
Facial expressions resulted in hyperalgesia in mice Construct to spontaneous head pain
of pain validi-
ty: +
MOH Rat Intraperitoneal; Mechanical Topiramate blocked the enhanced Fos Face No de novo headache prior [14–16, 18,
(sumatrip- 7–30 days hyperalgesia expression in the TNC and inhibited validi- to MOH 87]
tan and CSD-related cutaneous allodynia ty: ++ Allodynia is not equivalent
paraceta- bioelectrical Predictive to spontaneous head pain
mol) alterations validi-
Activation of ty: +
inflammatory Construct
markers in the validi-
TG ty: ?
Increased Fos
expression in
the TNC
Spontaneous Rat – Spontaneous Valproic acid prevented the Face Allodynia is not equivalent [59]
trigeminal episodic spontaneous changes in trigeminal validi- to spontaneous head pain
allodynia trigeminal allodynia ty: ++ Chronicity needs to be
allodynia Predictive defined
validi-
ty: +
Construct
validi-
ty: +
Monogenic FHM1 – Photophobia Rizatriptan reduced head grooming Face Monogenic form of [65]
migraine R192Q Unilateral head validi- migraine is not
mutation trans- grooming ty: ++ completely the same as
genic Lateralized Predictive common polygenic
mice winking/- validi- migraine
blinking ty: + Chronicity needs to be
Construct defined
validi-
ty: +++

Construct validity denotes comparable etiology. Face validity denotes analogy of symptoms. Predictive validity denotes responsiveness to migraine
treatment. Strong: +++, moderate: ++, mild: +, inconsistent: +/−, uncertain: ?
CGRP calcitonin gene-related peptide, MOH medication overuse headache, TG trigeminal ganglion, TNC trigeminal nucleus caudalis
Curr Pain Headache Rep (2018) 22:44 Page 5 of 8 44

suppressed, CSD susceptibility in mice [91]. Differences techniques to introduce multiple genetic variants identified through
among models, including those in animal species, kindling genome-wide association studies [98, 99]. Of course, the genesis
method, and pain surrogate, might contribute to the discrep- and chronification of migraine are not purely genetic; multiple
ancy of findings; however, the heterogeneity also suggests an factors should be considered simultaneously. Whether increased
imminent need for an ideal model of CM. Spontaneous tri- allostatic loads via the introduction of one or more migraine risk
geminal allodynia in rats [59] and transgenic mouse models of factors [100] increase the frequency of spontaneous attacks would
monogenic migraine [92] with spontaneous headache-like be- be of interest. An alternative approach might be to interrogate
haviors (such as those of mice carrying the human FHM1 specific nuclei or cellular groups via more physiological ap-
R192Q missense mutation in the Cacna1a gene [65]) is prob- proaches, such as optogenetics or chemogenetics, to see whether
ably more phenomenologically and pathophysiologically sim- the perturbation of neural circuits via these techniques could reca-
ilar to migraine. However, operational definitions of the fre- pitulate migraine phenomenology and whether repeated stimuli
quency and duration of spontaneous headache-like events could replicate specific features of CM.
might be required to characterize the extent to which these
models can represent the recalcitrant subtype of migraine.
Conclusions
Unsolved Problems and Future Directions This review raises critical points for the construction of an
for Animal Models of CM animal model of CM. We propose essential components for
a validated animal model of CM, describe the major contribu-
Drivers of Migraine Chronification tions and shortcomings of currently available models, and
highlight future directions for CM modeling in animal studies.
Sensitization of the central trigeminothalamic pathways may be The construction of a reliable animal model of CM is one step
an important cause of CM [5••, 93]. Such sensitization might toward the ultimate goal of developing preventive therapies to
occur during repeated migraine attacks through impaired de- halt progression from the episodic to the chronic state in
scending inhibition and/or enhanced descending facilitation of humans, as these models provide a platform for testing of
nociception [94, 95]. In addition, affective pain regions, such as the efficacy of potentially useful medications.
the insula and amygdala [11] or hypothalamus [96], might also
be important mediators in the chronification of migraine. Funding Acknowledgment This work was supported by Taipei
Elucidation of the roles of these structures during and following Veterans General Hospital (V106C-117 to S.P.C) and the Ministry
repetitive sensitization at the molecular, cellular, and circuit of Science and Technology of Taiwan (MOST 104-2314-B-075 -
levels might enable the construction of more ideal CM models 006 -MY3, 106-2321-B-010-009, and 107-2321-B-010-001 to
and generation of novel disease-modifying therapies. Given the S.P.C). The funders had no role in the study design, data collection
complex pathogenesis of CM, this disorder can be modeled and analysis, decision to publish, or preparation of the manuscript.
practically with a single phenotype or endophenotype serving
as a surrogate (as in most current models), as opposed to model- Compliance with Ethical Standards
ing of the entire phenotypic spectrum. Recently, two animal
models of MOH suggested a possible role of the amygdala in Conflict of Interest Tse-Ming Chou and Shih-Pin Chen declare no con-
the chronification of headache [90, 97]; these models are good flict of interest.
examples for further advancement of our knowledge.
Human and Animal Rights and Informed Consent This article does not
Nevertheless, given the shortcomings mentioned above, direct-
contain any studies with human or animal subjects performed by any of
ly inferring MOH models as CM models might not be correct. the authors.
Before the genesis of an ideal CM model, it is mandatory to
validate the findings using different model systems.
References
Spontaneous Nature of Migraine Attacks
Papers of particular interest, published recently, have been
The most currently available animal models are constructed using
highlighted as:
different kinds of repeated stimuli. To make the model convincing
• Of importance
and pathophysiologically closer to human migraine, spontaneous
•• Of major importance
attacks or more physiological triggers are needed. The use of
genetic animal models might be a good approach. To simulate 1. Natoli JL, Manack A, Dean B, Butler Q, Turkel CC, Stovner L,
common migraine, rather than rare monogenic forms of migraine et al. Global prevalence of chronic migraine: a systematic review.
[92], researchers might consider using modern gene-editing Cephalalgia. 2010;30(5):599–609.
44 Page 6 of 8 Curr Pain Headache Rep (2018) 22:44

2. Stark RJ, Ravishankar K, Siow HC, Lee KS, Pepperle R, Wang SJ. 23. Aurora SK, et al. Brainstem dysfunction in chronic migraine as
Chronic migraine and chronic daily headache in the Asia-Pacific evidenced by neurophysiological and positron emission tomogra-
region: a systematic review. Cephalalgia. 2013;33(4):266–83. phy studies. Headache. 2007;47(7):996–1003. discussion 1004–7
3. The International Classification of Headache Disorders, 3rd 24. Bergerot A, Holland PR, Akerman S, Bartsch T, Ahn AH,
edition. Cephalalgia. 2018;38(1):1–211. MaassenVanDenBrink A, et al. Animal models of migraine:
4. Lipton RB. Tracing transformation: chronic migraine classifica- looking at the component parts of a complex disorder. Eur J
tion, progression, and epidemiology. Neurology. 2009;72(5 Neurosci. 2006;24(6):1517–34.
Suppl):S3–7. 25. Jansen-Olesen I, Tfelt-Hansen P, Olesen J. Animal migraine
5.•• May A, Schulte LH. Chronic migraine: risk factors, mechanisms models for drug development: status and future perspectives.
and treatment. Nat Rev Neurol. 2016;12(8):455–64. A compre- CNS Drugs. 2013;27(12):1049–68.
hensive review for the clinical features and proposed pathophys- 26. Munro G, Jansen-Olesen I, Olesen J. Animal models of pain and
iology of chronic migraine migraine in drug discovery. Drug Discov Today. 2017;22(7):
6. Denuelle M, Fabre N, Payoux P, Chollet F, Geraud G. 1103–11.
Hypothalamic activation in spontaneous migraine attacks. 27. Storer RJ, Supronsinchai W, Srikiatkhachorn A. Animal models of
Headache. 2007;47(10):1418–26. chronic migraine. Curr Pain Headache Rep. 2015;19(1):467.
7. Welch KM, et al. Periaqueductal gray matter dysfunction in migraine: 28. Eikermann-Haerter K, Moskowitz MA. Animal models of migraine
cause or the burden of illness? Headache. 2001;41(7):629–37. headache and aura. Curr Opin Neurol. 2008;21(3):294–300.
8. Cernuda-Morollon E, et al. Increased VIP levels in peripheral 29. Chen SP, Ayata C. Novel therapeutic targets against spreading
blood outside migraine attacks as a potential biomarker of cranial depression. Headache. 2017;57(9):1340–58.
parasympathetic activation in chronic migraine. Cephalalgia. 30. Hoskin KL, Goadsby PJ. Comparison of more and less lipophilic
2015;35(4):310–6. serotonin (5HT1B/1D) agonists in a model of trigeminovascular
9. Cernuda-Morollon E, Larrosa D, Ramon C, Vega J, Martinez- nociception in cat. Exp Neurol. 1998;150(1):45–51.
Camblor P, Pascual J. Interictal increase of CGRP levels in pe- 31. Williamson DJ, Shepheard SL, Hill RG, Hargreaves RJ. The novel
ripheral blood as a biomarker for chronic migraine. Neurology. anti-migraine agent rizatriptan inhibits neurogenic dural vasodila-
2013;81(14):1191–6. tion and extravasation. Eur J Pharmacol. 1997;328(1):61–4.
10. Melo-Carrillo A, Lopez-Avila A. A chronic animal model of mi- 32. Goadsby PJ, Edvinsson L. Joint 1994 Wolff Award Presentation.
graine, induced by repeated meningeal nociception, characterized Peripheral and central trigeminovascular activation in cat is
by a behavioral and pharmacological approach. Cephalalgia. blocked by the serotonin (5HT)-1D receptor agonist 311C90.
2013;33(13):1096–105. Headache. 1994;34(7):394–9.
11. Schwedt TJ, Schlaggar BL, Mar S, Nolan T, Coalson RS, Nardos 33. Bigal ME, et al. Migraine in the triptan era: lessons from epide-
B, et al. Atypical resting-state functional connectivity of affective miology, pathophysiology, and clinical science. Headache.
pain regions in chronic migraine. Headache. 2013;53(5):737–51. 2009;49(Suppl 1):S21–33.
12. Coppola G, Iacovelli E, Bracaglia M, Serrao M, di Lorenzo C, 34. Zagami AS, Goadsby PJ, Edvinsson L. Stimulation of the superior
Pierelli F. Electrophysiological correlates of episodic migraine sagittal sinus in the cat causes release of vasoactive peptides.
chronification: evidence for thalamic involvement. J Headache Neuropeptides. 1990;16(2):69–75.
Pain. 2013;14:76. 35. Buzzi MG, Carter WB, Shimizu T, Heath H 3rd, Moskowitz
13. Burstein R, Jakubowski M, Garcia-Nicas E, Kainz V, Bajwa Z, MA. Dihydroergotamine and sumatriptan attenuate levels of
Hargreaves R, et al. Thalamic sensitization transforms localized CGRP in plasma in rat superior sagittal sinus during electrical
pain into widespread allodynia. Ann Neurol. 2010;68(1):81–91. stimulation of the trigeminal ganglion. Neuropharmacology.
14. De Felice M, et al. Triptan-induced latent sensitization: a possible 1991;30(11):1193–200.
basis for medication overuse headache. Ann Neurol. 2010;67(3): 36. Goadsby PJ, Edvinsson L. The trigeminovascular system and mi-
325–37. graine: studies characterizing cerebrovascular and neuropeptide
15. De Felice M, et al. Triptan-induced enhancement of neuronal nitric changes seen in humans and cats. Ann Neurol. 1993;33(1):48–56.
oxide synthase in trigeminal ganglion dural afferents underlies 37. Olesen J, Diener HC, Husstedt IW, Goadsby PJ, Hall D, Meier U,
increased responsiveness to potential migraine triggers. Brain. et al. Calcitonin gene-related peptide receptor antagonist BIBN
2010;133(8):2475–88. 4096 BS for the acute treatment of migraine. N Engl J Med.
16. Green AL, Gu P, de Felice M, Dodick D, Ossipov MH, Porreca F. 2004;350(11):1104–10.
Increased susceptibility to cortical spreading depression in an an- 38. Ho TW, Ferrari MD, Dodick DW, Galet V, Kost J, Fan X, et al.
imal model of medication-overuse headache. Cephalalgia. Efficacy and tolerability of MK-0974 (telcagepant), a new oral
2014;34(8):594–604. antagonist of calcitonin gene-related peptide receptor, compared
17. Bigal ME, Lipton RB. What predicts the change from episodic to with zolmitriptan for acute migraine: a randomised, placebo-con-
chronic migraine? Curr Opin Neurol. 2009;22(3):269–76. trolled, parallel-treatment trial. Lancet. 2008;372(9656):2115–23.
18. Supornsilpchai W, le Grand SM, Srikiatkhachorn A. Cortical hy- 39. Voss T, Lipton RB, Dodick DW, Dupre N, Ge JY, Bachman R,
perexcitability and mechanism of medication-overuse headache. et al. A phase IIb randomized, double-blind, placebo-controlled
Cephalalgia. 2010;30(9):1101–9. trial of ubrogepant for the acute treatment of migraine.
19. Schulte LH, Sprenger C, May A. Physiological brainstem mech- Cephalalgia. 2016;36(9):887–98.
anisms of trigeminal nociception: an fMRI study at 3T. 40. Dodick DW, Goadsby PJ, Silberstein SD, Lipton RB, Olesen J,
NeuroImage. 2016;124(Pt A):518–25. Ashina M, et al. Safety and efficacy of ALD403, an antibody to
20. Weiller C, May A, Limmroth V, Jüptner M, Kaube H, Schayck calcitonin gene-related peptide, for the prevention of frequent ep-
RV, et al. Brain stem activation in spontaneous human migraine isodic migraine: a randomised, double-blind, placebo-controlled,
attacks. Nat Med. 1995;1(7):658–60. exploratory phase 2 trial. Lancet Neurol. 2014;13(11):1100–7.
21. Akerman S, Holland PR, Goadsby PJ. Diencephalic and brainstem 41. Bigal ME, Edvinsson L, Rapoport AM, Lipton RB, Spierings
mechanisms in migraine. Nat Rev Neurosci. 2011;12(10):570–84. ELH, Diener HC, et al. Safety, tolerability, and efficacy of TEV-
22. Lai TH, Fuh JL, Lirng JF, Lin CP, Wang SJ. Brainstem 1H-MR 48125 for preventive treatment of chronic migraine: a multicentre,
spectroscopy in episodic and chronic migraine. J Headache Pain. randomised, double-blind, placebo-controlled, phase 2b study.
2012;13(8):645–51. Lancet Neurol. 2015;14(11):1091–100.
Curr Pain Headache Rep (2018) 22:44 Page 7 of 8 44

42. Bigal ME, Dodick DW, Rapoport AM, Silberstein SD, Ma Y, 61. De Felice M, et al. Capturing the aversive state of cephalic pain
Yang R, et al. Safety, tolerability, and efficacy of TEV-48125 for preclinically. Ann Neurol. 2013;74(2):257–65.
preventive treatment of high-frequency episodic migraine: a 62. Oshinsky ML, Gomonchareonsiri S. Episodic dural stimulation in
multicentre, randomised, double-blind, placebo-controlled, phase awake rats: a model for recurrent headache. Headache.
2b study. Lancet Neurol. 2015;14(11):1081–90. 2007;47(7):1026–36.
43. Sun H, Dodick DW, Silberstein S, Goadsby PJ, Reuter U, Ashina 63. Sufka KJ, Staszko SM, Johnson AP, Davis ME, Davis RE,
M, et al. Safety and efficacy of AMG 334 for prevention of epi- Smitherman TA. Clinically relevant behavioral endpoints in a re-
sodic migraine: a randomised, double-blind, placebo-controlled, current nitroglycerin migraine model in rats. J Headache Pain.
phase 2 trial. Lancet Neurol. 2016;15(4):382–90. 2016;17:40.
44. Dodick DW, Goadsby PJ, Spierings ELH, Scherer JC, Sweeney 64. Zhang M, Liu Y, Zhao M, Tang W, Wang X, Dong Z, et al.
SP, Grayzel DS. Safety and efficacy of LY2951742, a monoclonal Depression and anxiety behaviour in a rat model of chronic mi-
antibody to calcitonin gene-related peptide, for the prevention of graine. J Headache Pain. 2017;18(1):27.
migraine: a phase 2, randomised, double-blind, placebo-controlled 65. Chanda ML, Tuttle AH, Baran I, Atlin C, Guindi D, Hathaway G,
study. Lancet Neurol. 2014;13(9):885–92. et al. Behavioral evidence for photophobia and stress-related ipsi-
45.•• Silberstein SD, et al. Fremanezumab for the preventive treatment lateral head pain in transgenic Cacna1a mutant mice. Pain.
of chronic migraine. N Engl J Med. 2017;377(22):2113–22. A 2013;154(8):1254–62.
phase 3 trial showing that CGRP monoclonal antibody is effective 66. Baliki MN, Apkarian AV. Nociception, pain, negative moods, and
for chronic migraine, which might serve as a positive control to behavior selection. Neuron. 2015;87(3):474–91.
test the predictive validity of animal models 67. Kuner R, Flor H. Structural plasticity and reorganisation in chronic
46. Belzung C, Lemoine M. Criteria of validity for animal models of pain. Nat Rev Neurosci. 2016;18(1):20–30.
psychiatric disorders: focus on anxiety disorders and depression. 68. Dodick D, Silberstein S. Central sensitization theory of migraine:
Biol Mood Anxiety Disord. 2011;1(1):9. clinical implications. Headache. 2006;46(Suppl 4):S182–91.
47. Liu HY, Fuh JL, Lin YY, Chen WT, Wang SJ. Suicide risk in 69. Malick A, Burstein R. Peripheral and central sensitization during
patients with migraine and comorbid fibromyalgia. Neurology. migraine. Funct Neurol. 2000;15(Suppl 3):28–35.
2015;85(12):1017–23. 70. Dodick D, Silberstein S. Central sensitization theory of migraine:
48. Kao CH, Wang SJ, Tsai CF, Chen SP, Wang YF, Fuh JL. clinical implications. Headache: J Head Face Pain. 2006;46:S182–91.
Psychiatric comorbidities in allodynic migraineurs. Cephalalgia. 71. Pietrobon D, Moskowitz MA. Pathophysiology of migraine. Annu
2014;34(3):211–8. Rev Physiol. 2013;75:365–91.
49. Chen YC, Tang CH, Ng K, Wang SJ. Comorbidity profiles of 72. Goadsby PJ, Lipton RB, Ferrari MD. Migraine—current under-
chronic migraine sufferers in a national database in Taiwan. J standing and treatment. N Engl J Med. 2002;346(4):257–70.
Headache Pain. 2012;13(4):311–9. 73. Bolay H, Reuter U, Dunn AK, Huang Z, Boas DA, Moskowitz
50. Hamelsky SW, Lipton RB. Psychiatric comorbidity of migraine. MA. Intrinsic brain activity triggers trigeminal meningeal afferents
Headache. 2006;46(9):1327–33. in a migraine model. Nat Med. 2002;8(2):136–42.
51. Oh K, et al. Combination of anxiety and depression is associated 74. Burstein R, Yarnitsky D, Goor-Aryeh I, Ransil BJ, Bajwa ZH. An
with an increased headache frequency in migraineurs: a association between migraine and cutaneous allodynia. Ann
population-based study. BMC Neurol. 2014;14:238. Neurol. 2000;47(5):614–24.
52. Peterlin BL, Katsnelson MJ, Calhoun AH. The associations be- 75. Burstein R, Noseda R, Borsook D. Migraine: multiple processes,
tween migraine, unipolar psychiatric comorbidities, and stress- complex pathophysiology. J Neurosci. 2015;35(17):6619–29.
related disorders and the role of estrogen. Curr Pain Headache 76. Noseda R, Burstein R. Migraine pathophysiology: anatomy of the
Rep. 2009;13(5):404–12. trigeminovascular pathway and associated neurological symp-
53. Wang SJ, Chen PK, Fuh JL. Comorbidities of migraine. Front toms, cortical spreading depression, sensitization, and modulation
Neurol. 2010;1:16. of pain. Pain. 2013;154(Suppl 1):S44–53.
54. Bigal ME, Lipton RB. Overuse of acute migraine medications and 77. De Felice M, Ossipov MH, Porreca F. Update on medication-
migraine chronification. Curr Pain Headache Rep. 2009;13(4): overuse headache. Curr Pain Headache Rep. 2011;15(1):79–83.
301–7. 78. Silberstein SD, Lipton RB, Dodick DW, Freitag FG, Ramadan N,
55. Bigal ME, Serrano D, Buse D, Scher A, Stewart WF, Lipton RB. Mathew N, et al. Efficacy and safety of topiramate for the treat-
Acute migraine medications and evolution from episodic to chron- ment of chronic migraine: a randomized, double-blind, placebo-
ic migraine: a longitudinal population-based study. Headache. controlled trial. Headache. 2007;47(2):170–80.
2008;48(8):1157–68. 79. Whiteside GT, Adedoyin A, Leventhal L. Predictive validity of
56. Wang SJ, Fuh JL, Lu SR, Juang KD. Chronic daily headache in animal pain models? A comparison of the pharmacokinetic-
adolescents: prevalence, impact, and medication overuse. pharmacodynamic relationship for pain drugs in rats and humans.
Neurology. 2006;66(2):193–7. Neuropharmacology. 2008;54(5):767–75.
57. Ferrari LF, Levine JD, Green PG. Mechanisms mediating 80. Berge O-G. Predictive validity of behavioural animal models for
nitroglycerin-induced delayed-onset hyperalgesia in the rat. chronic pain. Br J Pharmacol. 2011;164(4):1195–206.
Neuroscience. 2016;317:121–9. 81. Rice AS, Cimino-Brown D, Eisenach JC, Kontinen VK, Lacroix-
58. Tipton AF, Tarash I, McGuire B, Charles A, Pradhan AA. The Fralish ML, Machin I, et al. Animal models and the prediction of
effects of acute and preventive migraine therapies in a mouse efficacy in clinical trials of analgesic drugs: a critical appraisal and
model of chronic migraine. Cephalalgia. 2016;36(11):1048–56. call for uniform reporting standards. Pain. 2008;139(2):243–7.
59. Oshinsky ML, Sanghvi MM, Maxwell CR, Gonzalez D, 82. Stucky NL, Gregory E, Winter MK, He YY, Hamilton ES,
Spangenberg RJ, Cooper M, et al. Spontaneous trigeminal McCarson KE, et al. Sex differences in behavior and expression
allodynia in rats: a model of primary headache. Headache. of CGRP-related genes in a rodent model of chronic migraine.
2012;52(9):1336–49. Headache. 2011;51(5):674–92.
60. Boyer N, Dallel R, Artola A, Monconduit L. General 83. Fried NT, et al. Region-specific disruption of the blood-brain bar-
trigeminospinal central sensitization and impaired descending rier following repeated inflammatory dural stimulation in a rat
pain inhibitory controls contribute to migraine progression. model of chronic trigeminal allodynia. Cephalalgia. 2018;38(4):
Pain. 2014;155(7):1196–205. 674–89.
44 Page 8 of 8 Curr Pain Headache Rep (2018) 22:44

84.• Pradhan AA, et al. Characterization of a novel model of chronic 91. Sukhotinsky I, Dilekoz E, Wang Y, Qin T, Eikermann-Haerter K,
migraine. Pain. 2014;155(2):269–74. The first mouse model Waeber C, et al. Chronic daily cortical spreading depressions sup-
showing that repetitive nitroglycerin infusion could elicit press spreading depression susceptibility. Cephalalgia.
prolonged mechanical hyperalgesia simulating the behavior dur- 2011;31(16):1601–8.
ing the chronification of migraine 92. Chen SP, Tolner EA, Eikermann-Haerter K. Animal models of
85. Kim SJ, et al. Differential development of facial and hind monogenic migraine. Cephalalgia. 2016;36(7):704–21.
paw allodynia in a nitroglycerin-induced mouse model of 93. Diener HC, Dodick DW, Goadsby PJ, Lipton RB, Olesen J,
chronic migraine; role of capsaicin sensitive primary affer- Silberstein SD. Chronic migraine—classification, characteristics
ents. Biol Pharm Bull. 2018;41(2):172–81. and treatment. Nat Rev Neurol. 2012;8(3):162–71.
86. Ben Aissa, M., et al., Soluble guanylyl cyclase is a critical regu- 94. Goadsby PJ, Hargreaves R. Refractory migraine and chronic mi-
lator of migraine-associated pain. Cephalalgia, 2017: p. graine: pathophysiological mechanisms. Headache. 2008;48(9):
333102417737778. 1399–405.
87. Yisarakun W, Chantong C, Supornsilpchai W, Thongtan T, 95. Goadsby PJ. Pathophysiology of migraine. Ann Indian Acad
Srikiatkhachorn A, Reuangwechvorachai P, et al. Up-regulation Neurol. 2012;15(Suppl 1):S15–22.
of calcitonin gene-related peptide in trigeminal ganglion follow-
96. Schulte LH, Allers A, May A. Hypothalamus as a mediator of
ing chronic exposure to paracetamol in a CSD migraine animal
chronic migraine: evidence from high-resolution fMRI.
model. Neuropeptides. 2015;51:9–16.
Neurology. 2017;88(21):2011–6.
88. Harris HM, Carpenter JM, Black JR, Smitherman TA, Sufka KJ.
The effects of repeated nitroglycerin administrations in rats; 97. Xie JY, de Felice M, Kopruszinski CM, Eyde N, LaVigne J,
modeling migraine-related endpoints and chronification. J Remeniuk B, et al. Kappa opioid receptor antagonists: a possible
Neurosci Methods. 2017;284:63–70. new class of therapeutics for migraine prevention. Cephalalgia.
89. Becerra L, Bishop J, Barmettler G, Xie Y, Navratilova E, Porreca 2017;37(8):780–94.
F, et al. Triptans disrupt brain networks and promote stress- 98. Anttila V, et al. Genome-wide meta-analysis identifies new sus-
induced CSD-like responses in cortical and subcortical areas. J ceptibility loci for migraine. Nat Genet. 2013;45(8):912–7.
Neurophysiol. 2016;115(1):208–17. 99. Chen, S.P., et al., Genome-wide association study identifies novel
90. Wanasuntronwong A, Jansri U, Srikiatkhachorn A. Neural hyper- susceptibility loci for migraine in Han Chinese resided in Taiwan.
activity in the amygdala induced by chronic treatment of rats with Cephalalgia, 2018;38(3):466–75.
analgesics may elucidate the mechanisms underlying psychiatric 100. Borsook D, Maleki N, Becerra L, McEwen B. Understanding
comorbidities associated with medication-overuse headache. migraine through the lens of maladaptive stress responses: a model
BMC Neurosci. 2017;18(1):1. disease of allostatic load. Neuron. 2012;73(2):219–34.

You might also like