You are on page 1of 11

The Journal of Neuroscience, April 29, 2015 • 35(17):6619 – 6629 • 6619

Disease Focus

Editor’s Note: Disease Focus articles provide brief overviews of a neural disease or syndrome, emphasizing potential links to basic
neural mechanisms. They are presented in the hope of helping researchers identify clinical implications of their research. For more
information, see http://www.jneurosci.org/misc/ifa_minireviews.dtl.

Migraine: Multiple Processes, Complex Pathophysiology


X Rami Burstein,1,3 Rodrigo Noseda,1,3 and David Borsook2,3
1Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, 2Department of

Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, Boston, Massachusetts 02115, and 3Harvard Medical School, Boston,
Massachusetts 02115

Migraine is a common, multifactorial, disabling, recurrent, hereditary neurovascular headache disorder. It usually strikes sufferers a few
times per year in childhood and then progresses to a few times per week in adulthood, particularly in females. Attacks often begin with
warning signs (prodromes) and aura (transient focal neurological symptoms) whose origin is thought to involve the hypothalamus,
brainstem, and cortex. Once the headache develops, it typically throbs, intensifies with an increase in intracranial pressure, and presents
itself in association with nausea, vomiting, and abnormal sensitivity to light, noise, and smell. It can also be accompanied by abnormal
skin sensitivity (allodynia) and muscle tenderness. Collectively, the symptoms that accompany migraine from the prodromal stage
through the headache phase suggest that multiple neuronal systems function abnormally. As a consequence of the disease itself or its
genetic underpinnings, the migraine brain is altered structurally and functionally. These molecular, anatomical, and functional abnor-
malities provide a neuronal substrate for an extreme sensitivity to fluctuations in homeostasis, a decreased ability to adapt, and the
recurrence of headache. Advances in understanding the genetic predisposition to migraine, and the discovery of multiple susceptible
gene variants (many of which encode proteins that participate in the regulation of glutamate neurotransmission and proper formation of
synaptic plasticity) define the most compelling hypothesis for the generalized neuronal hyperexcitability and the anatomical alterations
seen in the migraine brain. Regarding the headache pain itself, attempts to understand its unique qualities point to activation of the
trigeminovascular pathway as a prerequisite for explaining why the pain is restricted to the head, often affecting the periorbital area and
the eye, and intensifies when intracranial pressure increases.

Introduction such, is considered a genetic disorder may present as scintillating lights and scoto-
Migraine is a recurrent headache disorder (Ferrari et al., 2015). mas when affecting the visual cortex; pares-
affecting ⬃15% of the population during In some cases, the headache begins thesia, and numbness of the face and hands
the formative and most productive peri- with no warning signs and ends with when affecting the somatosensory cortex;
ods of their lives, between the ages of 22 sleep. In other cases, the headache may be tremor and unilateral muscle weakness
and 55 years (Stewart et al., 1994). It fre- preceded by a prodromal phase that in- when affecting the motor cortex or basal
quently starts in childhood, particularly cludes fatigue; euphoria; depression; irri- ganglia; and difficulty saying words (apha-
around puberty, and affects women more tability; food cravings; constipation; neck sia) when affecting the speech area (Cutrer
than men (3:1 female-to-male ratio; stiffness; increased yawning; and/or ab- and Olesen, 2006).
Leonardi et al., 2005; Bigal and Lipton, normal sensitivity to light, sound, and The pursuant headache is commonly
2009). It tends to run in families and, as smell (Kelman, 2004; Schoonman et al., unilateral, pulsating, aggravated by rou-
2006); and an aura phase that includes a tine physical activity, and can last a few
variety of focal cortically mediated neuro- hours to a few days (Headache Classifica-
logical symptoms that appear just before tion Committee of the International
Received Jan. 28, 2015; revised March 9, 2015; accepted March 20, 2015.
This research was supported by National Institutes of Health Grants and/or during the headache phase (Lashley, Headache Society, 2013). As the headache
NS-069847 (R.B.), NS-079687 (R.B.), NS-064050 (D.B.), NS-056195 (D.B.), 1941; Cutrer et al., 1998; Hansen et al., 2012, progresses, it may be accompanied by a
NS-073997 (D.B.), and AT-007530 (D.B.). 2013). Symptoms of migraine aura develop variety of autonomic symptoms (nausea,
The authors declare no competing financial interests. gradually, feature excitatory and inhibitory vomiting, nasal/sinus congestion, rhinor-
Correspondence should be addressed to Rami Burstein, CLS-649, 3
Blackfan Circle Boston, MA 02215. E-mail: rburstei@bidmc.harvard.edu.
phases, and resolve completely (Russell and rhea, lacrimation, ptosis, yawning, fre-
DOI:10.1523/JNEUROSCI.0373-15.2015 Olesen, 1996). Positive (gain-of-function) quent urination, and diarrhea), affective
Copyright © 2015 the authors 0270-6474/15/356619-11$15.00/0 and negative (loss-of-function) symptoms symptoms (depression and irritability),
6620 • J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 Burstein et al. • Migraine

cognitive symptoms (attention deficit, hyperexcitability (Ferrari et al., 2015). based on the following: (1) hypothalamic
difficulty finding words, transient amne- Mounting evidence for alterations in neurons are in a position to regulate the
sia, and reduced ability to navigate in brain structure and function that are sec- firing of preganglionic parasympathetic
familiar environments), and sensory ondary to the repetitive state of headache neurons in the superior salivatory nucleus
symptoms (photophobia, phonophobia, can explain the progression of disease (SSN) and sympathetic preganglionic
osmophobia, muscle tenderness, and cu- (Sprenger and Borsook, 2012). neurons in the spinal intermediolateral
taneous allodynia; Silberstein, 1995; Lip- nucleus (Hosoya et al., 1983, 1984; Tucker
ton et al., 2001; Kelman and Tanis, 2006). Prodromes and Saper, 1985; Loewy, 1990; Dampney,
The extent of these diverse symptoms In the context of migraine, prodromes are 2011; Fig. 1A); (2) the SSN can stimulate
suggests that migraine is more than a symptoms that precede the headache by the release of acetylcholine, vasoactive in-
headache. It is now viewed as a complex several hours. Examination of symptoms testinal peptide, and nitric oxide from
neurological disorder that affects multiple that are most commonly described by pa- meningeal terminals of postganglionic
cortical, subcortical, and brainstem areas tients point to the potential involvement parasympathetic neurons in the spheno-
that regulate autonomic, affective, cogni- of the hypothalamus (fatigue, depression, palatine ganglion (SPG), leading to dila-
tive, and sensory functions. As such, it is irritability, food cravings, and yawning), tion of intracranial blood vessels, plasma
evident that the migraine brain differs brainstem (muscle tenderness and neck protein extravasation, and local release of
from the nonmigraine brain (Borsook et stiffness), cortex (abnormal sensitivity to inflammatory molecules capable of acti-
al., 2012a) and that an effort to unravel the light, sound, and smell), and limbic sys- vating pial and dural branches of menin-
pathophysiology of migraine must ex- tem (depression and anhedonia) in the geal nociceptors; (3) meningeal blood
pand beyond the simplistic view that there prodromal phase of a migraine attack vessels are densely innervated by para-
are “migraine generator” areas (Borsook (Maniyar et al., 2014). Given that symp- sympathetic fibers (Larsson et al., 1976;
and Burstein, 2012). In studying migraine toms such as fatigue, yawning, food crav- Nozaki et al., 1993; Suzuki and Hardebo,
pathophysiology, we must consider how ing, and transient mood changes occur 1993); (4) activation of SSN neurons can
different neural networks interact with naturally in all humans, it is critical that modulate the activity of central trigemi-
each other to allow migraine to com- we understand how their occurrence novascular neurons in the spinal trigemi-
mence with stressors such as insufficient triggers a headache; whether the routine nal nucleus (SpV; Akerman et al., 2012);
sleep, skipping meals, stressful or post- occurrence of these symptoms in mi- (5) activation of meningeal nociceptors
stressful periods, hormonal fluctuations, graineurs (i.e., when no headache devel- appears to depend partially on enhanced
alcohol, certain foods, flickering lights, ops) differs mechanistically from their activity in the SPG (Bolay et al., 2002); (6)
noise, or certain scents, and why migraine occurrence before the onset of migraine; enhanced cranial parasympathetic tone
attacks are sometimes initiated by these and why yawning, food craving, and fa- during migraine is evident by lacrimation
triggers and sometimes not. We must tigue do not trigger a migraine in healthy and nasal congestion (Liveing, 1873;
tackle the enigma of how attacks are re- subjects. Recently, much attention has Havanka-Kanniainen et al., 1988; Shech-
solved on their own or just weaken and been given to the hypothalamus because it ter et al., 2002); and, finally, (7) blockade
become bearable by sleep, relaxation, plays a key role in many aspects of human of the sphenopalatine ganglion provides
food, and/or darkness. We must explore circadian rhythms (wake-sleep cycle, partial or complete relief of migraine pain
the mechanisms by which the frequency body temperature, food intake, and hor- (Sluder, 1908; Kudrow, 1980; Diamond
of episodic migraine increases over time monal fluctuations) and in the continu- and Dalessio, 1982; Waldman, 1993; Kud-
(from monthly to weekly to daily), and ous effort to maintain homeostasis. row et al., 1995; Maizels et al., 1996; Yar-
why progression from episodic to chronic Because the migraine brain is extremely nitsky et al., 2003).
migraine is uncommon. sensitive to deviations from homeostasis, The second proposal suggests that
it seems reasonable that hypothalamic hypothalamic and brainstem neurons
Disease mechanisms neurons that regulate homeostasis and that regulate responses to deviation
In many cases, migraine attacks are likely circadian cycles are at the origin of some from physiological and emotional ho-
to begin centrally, in brain areas capable of the migraine prodromes. meostasis can lower the threshold for
of generating the classical neurological Unraveling the mechanisms by which the transmission of nociceptive trigemi-
symptoms of prodromes and aura, hypothalamic and brainstem neurons can novascular signals from the thalamus to
whereas the headache phase begins with trigger a headache is central to our ability the cortex—a critical step in establishing
consequential activation of meningeal no- to develop therapies that can intercept the the headache experience (Noseda et al.,
ciceptors at the origin of the trigemino- headache during the prodromal phase 2014). This proposal is based on under-
vascular system (Noseda and Burstein, (i.e., before the headache begins; Géraud standing how the thalamus selects, ampli-
2013). While some clues about how the and Donnet, 2013). The ongoing effort to fies, and prioritizes information it
occurrence of aura can activate nocicep- answer this question focuses on two very eventually transfers to the cortex (McCor-
tors in the meninges exist, nothing is different possibilities (Fig. 1). The first mick, 1992; Sherman and Guillery, 1998;
known about the mechanisms by which suggests that hypothalamic neurons that Sherman, 2005), and how hypothalamic
common prodromes initiate the headache respond to changes in physiological and and brainstem nuclei regulate relay
phase or what sequence of events they emotional homeostasis can activate men- thalamocortical neurons (Saper et al.,
trigger that results in activation of the ingeal nociceptors by altering the balance 2005, 2010). It is constructed from recent
meningeal nociceptors. A mechanistic between parasympathetic and sympa- evidence that relay trigeminothalamic
search for a common denominator in mi- thetic tone in the meninges (Burstein and neurons in sensory thalamic nuclei re-
graine symptomatology and characteris- Jakubowski, 2005) toward the predomi- ceive direct input from hypothalamic
tics points heavily toward a genetic nance of parasympathetic tone (Shechter neurons that contain dopamine, hista-
predisposition to generalized neuronal et al., 2002). Support for such a proposal is mine, orexin, and melanin concentrating
Burstein et al. • Migraine J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 • 6621

Figure 1. From prodromes to headache: proposed hypothesis for the initiation of headache by the hypothalamus and brainstem. A, Hypothalamic–parasympathetic pathway for the
activation of meningeal nociceptors by neurons that regulate homeostasis, circadian rhythms, and autonomic functions (adapted from Burstein and Jakubowski, 2005). Hypothalamically
mediated activation of preganglionic parasympathetic neurons in the SSN can trigger the release of acetylcholine, vasoactive intestinal peptide, and nitric oxide from meningeal
terminals of postganglionic parasympathetic neurons in the SPG. B, C, Neurochemical pathways capable of modulating the excitability of relay thalamocortical neurons in response to
deviation from physiological (food intake, sleep) and emotional (stress, anxiety) homeostasis. The illustration (top right) shows the hypothalamic and brainstem origin of each of the
pathways found to converge on thalamic trigeminovascular neurons (adapted from Kagan et al., 2013; Noseda et al., 2014). The photomicrographs show the extent of innervation by
vesicular glutamate transporter, vesicular GABA transporter, serotonin transporter, dopamine beta hydroxylase, tyrosine hydroxylase, histamine, melanin-concentrating hormone, and
orexin A. D, Conceptual illustration of how brainstem tone (allostatic load) may allow the headache to develop incosistently in response to identical changes in external and internal
conditions. Brainstem “state of tone” can limit afferent nociceptive drive in migraine-susceptible individuals (adapted from Borsook and Burstein, 2012). Fluctuation of activity in
brainstem neurons is thought to drive adaptive behavior. In the context of migraine, this can apply to the modulation of nociceptive signals from the meninges. The gating of these signals
depends on the threshold of the neural networks that modify these afferent signals. Thus, the robustness of the “gate” that allows nociceptive signals to drive central trigeminovascular
neurons (and thus headache) is dictated by brainstem tone. When the brainstem tone is high [red dot below line of migraine threshold (MT)], nociceptive signals are inhibited; and when
the brainstem tone is low (red dot above MT), afferent signals are not effectively blocked. The model illustrates the following three functional brainstem states: (1) normal state, when
cyclical brainstem activity is high, the potency of pain facilitation (enhanced synaptic strength in the dorsal horn) is too high to allow nociceptive signals from the periphery to drive the
central neurons into the active state (left); (2) threshold state, at threshold, the system has reached a primed state that could tip into a functional state that would allow nociceptive drive
from the dura to activate the central trigeminovascular neurons (middle); and (3) migraine state, when cyclical brainstem activity is low (more sensitive to stimuli), nociceptive signals
from the periphery can drive the central neurons into the active state (right). A11, Hypothalamic dopaminergic nucleus; C1/C2, cervical spinal cord segments; DR, dorsal raphe nucleus;
DTM, dorsal tuberomammary hypothalamic nucleus; LC, locus ceruleus; LH, lateral hypothalamus, PeF, perifornical area; RMg, nucleus raphe magnus; TG, trigeminal ganglion.
6622 • J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 Burstein et al. • Migraine

hormone (MCH), and brainstem neurons genes involved in inflammatory process- headache itself (Burstein and Jakubowski,
that contain noradrenaline and serotonin ing; and a host of changes in cortical per- 2005). Additional projections of trigemino-
(Noseda et al., 2014; Fig. 1 B, C). In prin- fusion and enzymatic activity that include vascular SpV neurons are found in the
ciple, each of these neuropeptides/neu- opening of the megachannel Panx1, acti- thalamic ventral posteromedial (VPM),
rotransmitters can shift the activity of vation of caspase-1, and a breakdown of posterior (PO), and parafascicular nuclei
thalamic neurons from burst to tonic the blood– brain barrier (Karatas et al., (Malick et al., 2000). Relay trigeminovascu-
mode if it is excitatory (dopamine, and 2013). Outside the brain, caspase-1 acti- lar thalamic neurons that project to the
high concentration of serotonin, nor- vation can initiate inflammation by re- somatosensory, insular, motor, parietal as-
adrenaline, histamine, orexin), and from leasing high-mobility group protein B1 sociation, retrosplenial, auditory, visual,
tonic to burst mode if it is inhibitory and interleukin-1␤ into the CSF, which and olfactory cortices are in a position to
(MCH and low concentration of sero- then activates nuclear factor-␬B in astro- construct the specific nature of migraine
tonin). The opposing factors that regulate cytes, with the consequential release of pain (i.e., location, intensity, and quality)
the firing of relay trigeminovascular tha- cyclooxygenase-2 and inducible nitric ox- and many of the cortically mediated symp-
lamic neurons provide an anatomical ide synthase (iNOS) into the subarach- toms that distinguish between migraine
foundation for explaining why pro- noid space (Karatas et al., 2013). The headache and other pains. These include
dromes give rise to some migraine attacks introduction into the meninges of these transient symptoms of motor clumsiness,
but not to others, and why external (e.g., proinflammatory molecules, as well as difficulty focusing, amnesia, allodynia, pho-
exposure to strong perfume) and internal calcitonin gene-related peptide (CGRP) nophobia, photophobia, and osmophobia
conditions (e.g., skipping a meal and feel- and nitric oxide (Wahl et al., 1994; Ob- (Noseda et al., 2011). Figure 2A illustrates
ing hungry, sleeping too little and being renovitch et al., 2002; Russo, 2015), may the complexity of the trigeminovascular
tired, or simple stress) trigger migraine at- be the link between aura and headache be- pathway.
tacks so inconsistently. In the context of cause the meninges are densely innervated Activation. Studies in animals show
migraine, the convergence of these hypo- by pain fibers whose activation distin- that CSD initiates delayed activation (Fig.
thalamic and brainstem neurons on tha- guishes headaches of intracranial origin 2 B, C) and immediate activation (Fig. 2D)
lamic trigeminovascular neurons can (e.g., migraine, meningitis, and subarach- of peripheral and central trigeminovascu-
establish high and low set points for the noid bleeds) from headaches of extracra- lar neurons in a fashion that resembles the
allostatic load of the migraine brain (Bor- nial origin (e.g., tension-type headache, classic delay and occasional immediate
sook et al., 2012b). The allostatic load, de- cervicogenic headache, or headaches onset of headache after aura (Zhang et al.,
fined as the amount of brain activity caused by mild trauma to the cranium). 2010, 2011), and that systemic adminis-
required to appropriately manage the tration of the M-type potassium channel
level of emotional or physiological stress Anatomy and physiology of the opener KCNQ2/3 can prevent the CSD-
at any given time (McEwen, 1998, 2004; trigeminovascular pathway: from induced activation of the nociceptors
McEwen and Wingfield, 2003; Peters and activation to sensitization (Zhang et al., 2013). These findings sup-
McEwen, 2012), can explain why external Anatomical description. The trigemino- port the notion that the onset of the head-
and internal conditions only trigger head- vascular pathway conveys nociceptive in- ache phase of migraine with aura
ache some of the times, when they coin- formation from the meninges to the coincides with the activation of meningeal
cide with the right circadian phase of brain. The pathway originates in trigemi- nociceptors at the peripheral origin of the
cyclic rhythmicity of brainstem, and hy- nal ganglion neurons whose peripheral trigeminovascular pathway. Whereas the
pothalamic and thalamic neurons that axons reach the pia, dura, and large cere- vascular, cellular, and molecular events
preserve homeostasis (Fig. 1D). bral arteries (Uddman et al., 1985), and involved in the activation of meningeal
whose central axons reach the nociceptive nociceptors by CSD are not well under-
Cortical spreading depression dorsal horn laminae of the SpV (Liu et al., stood, a large body of data suggests that
Clinical and preclinical studies suggest 2004). In the SpV, the nociceptors con- transient constriction and dilatation of
that migraine aura is caused by cortical verge on neurons that receive additional pial arteries and the development of dural
spreading depression (CSD), a slowly input from the periorbital skin and peri- plasma protein extravasation, neurogenic
propagating wave of depolarization/exci- cranial muscles (Davis and Dostrovsky, inflammation, platelet aggregation, and
tation followed by hyperpolarization/in- 1988). The ascending axonal projections mast cell degranulation (Moskowitz and
hibition in cortical neurons and glia of trigeminovascular SpV neurons trans- Macfarlane, 1993; Moskowitz, 1993), many
(Leao, 1944; Sugaya et al., 1975; Cutrer et mit monosynaptic nociceptive signals to of which may be driven by CSD-dependent
al., 1998; Hadjikhani et al., 2001). While (1) brainstem nuclei, such as the ventro- peripheral CGRP release (Russo, 2015),
specific processes that initiate CSD in hu- lateral periaqueductal gray, reticular for- can introduce to the meninges proinflam-
mans are not known, mechanisms that in- mation, superior salivatory, parabrachial, matory molecules, such as histamine, bra-
voke inflammatory molecules as a result cuneiform, and the nucleus of the solitary dykinin, serotonin, and prostaglandins
of emotional or physiological stress, such tract; (2) hypothalamic nuclei, such as the (prostaglandin E2), and a high level of hy-
as lack of sleep, may play a role. In the anterior, lateral, perifornical, dorsome- drogen ions-thus altering the molecular en-
cortex, the initial membrane depolariza- dial, suprachiasmatic, and supraoptic; vironment in which meningeal nociceptors
tion is associated with a large efflux of po- and (3) basal ganglia nuclei, such as the exist.
tassium; influx of sodium and calcium; caudate-putamen, globus pallidus, and sub- Sensitization. When activated in the al-
release of glutamate, ATP, and hydrogen stantia innominata (Malick et al., 2000). tered molecular environment described
ions; neuronal swelling (Hansen and Zeu- These projections may be critical for the ini- above, peripheral trigeminovascular neu-
then, 1981; Mutch and Hansen, 1984; tiation of nausea, vomiting, yawning, lacri- rons become sensitized (their response
Schock et al., 2007; Charles and Brennan, mation, urination, loss of appetite, fatigue, threshold decreases and their response
2009; Chang et al., 2010); upregulation of anxiety, irritability, and depression by the magnitude increases) and begin to re-
Burstein et al. • Migraine J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 • 6623

Figure 2. Activation and sensitization of the trigeminovascular pathway provide anatomical and physiological substrates for migraine headache and its associated symptoms. A, Intricate
anatomy of the trigeminovascular pathway. B–D, Single-unit recording showing delayed (B, C) and immediate (D) activation of a meningeal nociceptor (B) and two SpV neurons (C, D) following
the induction of CSD in the visual cortex of the rat. E, Activation and sensitization of a meningeal nociceptor. Baseline responses to mechanical stimulation of the dura (blue) increase (red) after
exposure to inflammatory soup (IS). F, Activation and sensitization of a trigeminovascular neuron in the SpV. Baseline responses to mechanical stimulation of the periorbital skin (blue) increased
(red) after a brief exposure of the dura to IS. G, Activation and sensitization of thalamic trigeminovascular neurons. Baseline responses to mechanical stimulation of the lower limb (blue) increased
in magnitude and duration (red) following brief application of IS to the dura. H, Contrast analysis of BOLD signals registered in fMRI scans of the human trigeminal ganglion during migraine attacks.
I, Contrast analysis of BOLD signals registered in fMRI scans of the human SpV following innocuous mechanical stimulation of the periorbital skin during migraine. J, Contrast analysis of BOLD signals
registered in fMRI scans of the human thalamus following innocuous mechanical stimulation of the skin on the dorsum of the hand during migraine. Red/yellow area depicts the periorbital area of
referred pain. Purple/yellow areas depict region of cephalic and extracephalic allodynia. Au, Auditory cortex; C6 –C7, sixth and seventh spinal cord segments; DRG, dorsal root ganglion; Ins, insular
cortex; Ect, ectorhinal cortex; LP, lateral posterior thalamic nucleus; M1/M2, primary and secondary motor cortices; PAG, periaqueductal gray; PB, parabrachial nucleus; PtA, parietal association
cortex; Pul, pulvinar; RS, retrosplenial cortex; S1/S2, primary and secondary somatosensory cortices; TG, trigeminal ganglion; V1/V2, primary and secondary visual cortex. Parts of this figure were
adapted from Strassman et al., 1996; Burstein et al., 1998, 2010; and Zhang et al., 2010 and 2011.
6624 • J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 Burstein et al. • Migraine

spond to dura stimuli to which they headache. Further support for this con- receptor, which in turn may amplify and re-
showed minimal or no response at base- cept was provided recently by studies inforce pain transmission, and the develop-
line (Strassman et al., 1996; Fig. 2E). showing that humanized monoclonal an- ment of allodynia and central sensitization
When central trigeminovascular neurons tibodies against CGRP, molecules that are (Burstein et al., 2000). Network-wise, wide-
in laminae I and V of SpV (Fig. 2F ) and in too big to penetrate the blood– brain bar- spread neuronal hyperexcitability may also
the thalamic PO/VPM nuclei (Fig. 2G) be- rier and act centrally (according to the be driven by thalamocortical dysrhythmia
come sensitized, their spontaneous activ- companies that developed them), are ef- (Llinás, 1988; Steriade and Llinás, 1988;
ity increases, their receptive fields expand, fective in preventing migraine (Diener, Steriade et al., 1993; Fuggetta and Noh,
and they begin to respond to innocuous 2014; Dodick et al., 2014a,b; Russo, 2015). 2013), defective modulatory brainstem
mechanical and thermal stimulation of Along this line, it was also reported that circuits that regulate excitability at multi-
cephalic and extracephalic skin areas as if drugs that act on central trigeminovascu- ple levels along the neuraxis (Bahra et al.,
it were noxious (Burstein et al., 1998, lar neurons [e.g., dihydroergotamine 2001); and inherently improper regula-
2010). The human correlates of the elec- (DHE)] are equally effective in reversing tion/habituation of cortical (Coppola and
trophysiological measures of neuronal an already developed central sensitization Schoenen, 2012; Coppola et al., 2013),
sensitization in animal studies are evident (Pozo-Rosich and Oshinsky, 2005)—a thalamic (Burstein et al., 2010), and
in contrast analysis of BOLD signals reg- possible explanation for DHE effective- brainstem (Weiller et al., 1995; Moulton
istered in fMRI scans of the human tri- ness in aborting migraine after the failure et al., 2008) functions by limbic structures,
geminal ganglion (Fig. 2H ), spinal of therapy with triptans. such as the hypothalamus, amygdala, nu-
trigeminal nucleus (Fig. 2I ), and the thal- cleus accumbens, caudate, putamen, and
amus (Fig. 2J ), all measured during mi- Genetics and the hyperexcitable brain globus pallidus. Given that 2 of the 13 sus-
graine attacks. The clinical manifestation Family history points to a genetic predis- ceptibility genes regulate synaptic devel-
of peripheral sensitization during mi- position to migraine. A genetic associa- opment and plasticity, it is reasonable to
graine, which takes ⬃10 min to develop, tion with migraine was first observed and speculate that some of the networks men-
includes the perception of throbbing defined in patients with familial hemiple- tioned above may not be properly wired to
headache and the transient intensification gic migraine (FHM). The three genes set a normal level of habituation through-
of headache while bending over or cough- identified with FHM encode proteins that out the brain, thus explaining the multi-
ing, activities that momentarily increase regulate glutamate availability in the syn- factorial nature of migraine. Along this
intracranial pressure (Blau and Dexter, apse. FHM1 (CACNA1A) encodes the line, it is also tempting to propose that at
1981). The clinical manifestation of sensi- pore-forming ␣1 subunit of the P/Q type least some of the structural alterations
tization of central trigeminovascular neu- calcium channel (Ophoff et al., 1996; Du- seen in the migraine brain may be inher-
rons in the SpV, which takes 30 – 60 min cros et al., 2001); FHM2 (ATP1A2) ited and, as such, may be the “cause” of
to develop and 120 min to reach full ex- encodes the ␣2 subunit of the Na ⫹/K ⫹- migraine, rather than being secondary to
tent, include the development of cephalic ATPase pump (De Fusco et al., 2003); and (i.e., being caused by) the repeated head-
allodynia signs such as scalp and muscle the FHM3 (SCN1A) encodes the ␣1 sub- ache attacks. But this concept awaits
tenderness and hypersensitivity to touch unit of the neuronal voltage-gated Nav1.1 evidence.
(Burstein et al., 2000; Bigal et al., 2008; channel (Dichgans et al., 2005). Collec-
Lipton et al., 2008). These signs are often tively, these genes regulate transmitter Structural and functional brain
recognized in patients reporting that they release, glial ability to clear (reuptake) alterations
avoid wearing glasses, earrings, hats, or glutamate from the synapse, and the gen- Brain alterations can be categorized into
any other object that come in contact with eration of action potentials (Ferrari et al., the following two processes: (1) alteration
the facial skin during migraine (Burstein 2015). Since these early findings, large in brain function and (2) alterations in
et al., 2000; Bigal et al., 2008; Lipton et al., genome-wide association studies have identi- brain structure (Fig. 3). Functionally, a
2008). The clinical manifestation of tha- fied 13 susceptibility gene variants for mi- variety of imaging techniques used to
lamic sensitization during migraine, graine with and without aura (Anttila et al., measure relative activation in different
which takes 2– 4 h to develop, also in- 2010, 2013; Chasman et al., 2011; Freilinger brain areas in migraineurs (vs control
cludes extracephalic allodynia signs that et al., 2012), three of which regulate glutamin- subjects) revealed enhanced activation in
cause patients to remove tight cloth and ergic neurotransmission (MTDH/AEG-1 the periaqueductal gray (Weiller et al.,
jewelry, and avoid being touched, mas- downregulates glutamate transporter, LPR1 1995); red nucleus and substantia nigra
saged, or hugged (Burstein et al., 2000; Bi- modulates synaptic transmission through the (Cao et al., 2002); hypothalamus (Denu-
gal et al., 2008; Lipton et al., 2008). NMDA receptor, and MEF-2D regulates the elle et al., 2007); posterior thalamus (Bur-
Evidence that triptans, 5HT1B/1D agonists glutamatergic excitatory synapse), and two of stein et al., 2010); cerebellum, insula,
that disrupt communications between pe- which regulate synaptic development and cingulate and prefrontal cortices, anterior
ripheral and central trigeminovascular plasticity (ASTN2 is involved in the structural temporal pole, and the hippocampus (Af-
neurons in the dorsal horn (Levy et al., development of cortical layers, and FHL5 reg- ridi et al., 2005; Moulton et al., 2011); and
2004), are more effective in aborting mi- ulates cAMP-sensitive CREB proteins in- decreased activation in the somatosensory
graine when administered early (i.e., be- volvedinsynapticplasticity;Anttilaetal.,2010, cortex (Tessitore et al., 2011), nucleus cu-
fore the development of central 2013; Chasman et al., 2011; Freilinger et al., neiformis (Moulton et al., 2008), caudate,
sensitization and allodynia) rather than 2012). These findings provide the most plau- putamen, and pallidum (Maleki et al.,
late (i.e., after the development of allo- sible explanation for the “generalized” neuro- 2011b). All of these activity changes oc-
dynia; Burstein and Jakubowski, 2004; nal hyperexcitability of the migraine brain. curred in response to nonrepetitive stim-
Burstein et al., 2004) provides further In the context of migraine, increased uli, and in the cingulate and prefrontal
support for the notion that meningeal no- activity in glutamatergic systems can lead cortex they occurred in response to repet-
ciceptors drive the initial phase of the to excessive occupation of the NMDA itive stimuli (Aderjan et al., 2010). Collec-
Burstein et al. • Migraine J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 • 6625

Figure 3. Functional (MRIFunct) and morphometric (MRIMorph) changes in the migraine brain. The examples illustrate changes in functional and morphological measures in cortical (somatosen-
sory) and subcortical (basal ganglia) areas, as well as sex differences in male and female migraineurs. Bottom left conceptualizes dendritic tree density (blue ⫽ volume loss; red ⫽ volume gain).
Somatosensory cortex: increased somatosensory activation to a noxious stimulus (pain threshold ⫹1C) applied to the face (forehead) and increased cortical thickness in episodic migraineur. MRIFunct
shows bilateral activation in the primary somatosensory cortex (top; yellow-orange). MRIMorph shows significant changes in cortical volume (green) in high vs low episodic migraineurs vs healthy
control subjects. Basal ganglia: decreased activation (top) in the caudate (MRIFunct) in high-frequency vs low-frequency episodic migraineurs in response to a noxious heat stimulus (pain threshold
⫹1C) is associated with increased volume (MRIMorph) in the structure (bottom). Sex differences: overlap of disease-related and sex-related functional differences (MRIFunct) in men vs women (top;
orange-red) showing decreased activation in episodic migraineurs to a noxious stimulus (pain threshold ⫹1C) applied to the face (forehead). Also shown are decreased activations in female episodic
migraineurs (FM) vs healthy control subjects (C). Bottom shows increased cortical thickness in female migraineurs (FM) vs male migraineurs (MM) in the insula. Other areas showing a similar sex
difference included the precuneus. The issue of sex-related changes in the migraine brain have been reviewed previously (Borsook et al., 2014). Parts of this figure are adapted from Maleki et al.,
2011b, 2012a,b; and Borsook et al., 2013.

tively, these studies support the concept cortical and subcortical structures may inferior temporal cortex in osteoarthritis pa-
that the migraine brain lacks the ability to also depend on the frequency of migraine tients with chronic back pain (Apkarian et al.,
habituate itself and consequently becomes attacks for a number of cortical (Maleki et 2004; Baliki et al., 2011). Whereas some of the
hyperexcitable (Coppola et al., 2007, al., 2011a, 2013) and subcortical regions brain alterations seen in migraineurs depend
2009). It is a matter of debate, however, if (Maleki et al., 2012b). As discussed above, on the sex of the patient (Maleki et al., 2012b;
such changes are unique to migraine it is unclear whether such changes are ge- Borsook et al., 2014), little can be said about
headache. Evidence for nearly identical netically predetermined or simply a result the role played by the sex of patients who ex-
activation patterns in other pain condi- of the repetitive exposure to pain/stress. perience other pain conditions.
tions, such as low back pain, neuropathic Favoring the latter are studies showing
pain, fibromyalgia, irritable bowel syn- that similar gray matter changes occur-
drome, and cardiac pain (Apkarian, 2008; ring in patients experiencing other Treatments in development
Baliki et al., 2008), raises the possibility chronic pain conditions (Apkarian et al., Migraine therapy has two goals: to termi-
that differences between somatic pain and 2004; Baliki et al., 2011) are reversible and nate acute attacks; and to prevent the next
migraine pain are not due to differences in that the magnitude of these changes can attack from happening. The latter can po-
central pain processing. be correlated with the duration of disease tentially prevent the progression from ep-
Anatomically, voxel-based morphome- (Rodriguez-Raecke et al., 2009). Further isodic to chronic state. Regarding the
try and diffusion tensor imaging studies in complicating our ability to determine effort to terminate acute attacks, migraine
migraine patients (vs control subjects) have how the migraine brain differs from the represents one of the few pain conditions
revealed thickening of the somatosensory brain of a patient experiencing other for which a specific drug (i.e., triptan) has
cortex (DaSilva et al., 2007; Hadjikhani, chronic pain conditions are anatomical been developed based on understanding
2008; Maleki et al., 2012a); increased gray findings showing decreased gray matter the mechanisms of the disease. In contrast,
matter density in the caudate (Maleki et al., density in the prefrontal cortex, thalamus, the effort to prevent migraine from happen-
2011b); and gray matter volume loss in the posterior insula, secondary somatosensory ing is likely to face a much larger challenge
superior temporal gyrus, inferior frontal cortex, precentral and postcentral gyrus, given that migraine can originate in an un-
gyrus, precentral gyrus, anterior cingulate hippocampus, and temporal pole of chronic known number of brain areas (see above),
cortex, amygdala, parietal operculum, back pain patients; anterior insula and or- and is associated with generalized functional
middle and inferior frontal gyrus, inferior bitofrontal cortex of complex regional pain and structural brain abnormalities. A num-
frontal gyrus, and bilateral insula (DaSilva syndrome patients; and the insula, mid- ber of treatments that attract attention are
et al., 2007; Valfrè et al., 2008). Changes in anterior cingulate cortex, hippocampus, and briefly reviewed below.
6626 • J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 Burstein et al. • Migraine

Medications Conclusions Bahra A, Matharu MS, Buchel C, Frackowiak RS,


The most exciting drug currently under Migraine is a common and undertreated Goadsby PJ (2001) Brainstem activation
development is humanized monoclonal specific to migraine headache. Lancet 357:
disease. For those who suffer, it is a major
1016 –1017. CrossRef Medline
antibodies against CGRP. The develop- cause of disability, including missing Baliki MN, Geha PY, Apkarian AV, Chialvo DR
ment of these monoclonal antibodies work or school, and it frequently has asso- (2008) Beyond feeling: chronic pain hurts the
are directed at both CGRP and its recep- ciated comorbidities such as anxiety and brain, disrupting the default-mode network
tors (Diener, 2014; Bigal et al., 2015; depression. To put this in context, it is a dynamics. J Neurosci 28:1398 –1403. CrossRef
Edvinsson, 2015; Russo, 2015). The concept is leading cause of suicide, an indisputable Medline
proof of the severity of the distress that the Baliki MN, Schnitzer TJ, Bauer WR, Apkarian AV
based on CGRP localization in the trigeminal
(2011) Brain morphological signatures for
ganglion and its relevance to migraine patho- disease may inflict on the individual
chronic pain. PLoS One 6:e26010. CrossRef
physiology (Russo, 2015). In recent phase (Wang et al., 2009; Fuller-Thomson et al., Medline
II randomized placebo-controlled trials 2013). There is currently no objective di- Bigal ME, Lipton RB (2009) The epidemiology,
(clinical trials NCT0177252, NCT01625988, agnosis or treatment that is universally ef- burden, and comorbidities of migraine. Neu-
NCT02025556, and NCT01723514; clini- fective in aborting or preventing attacks. rol Clin 27:321–334. CrossRef Medline
As an intermittent disorder, migraine rep- Bigal ME, Ashina S, Burstein R, Reed ML, Buse D,
caltrials.gov), the neutralizing humanized Serrano D, Lipton RB (2008) Prevalence and
monoclonal antibodies against CGRP resents a neurological condition wherein
characteristics of allodynia in headache suffer-
systems that continuously evaluate errors
(ALD403, LY2951742, LBR-101) adminis- ers: a population study. Neurology 70:1525–
(error detection) frequently fail, thus add- 1533. CrossRef Medline
tered by injection for the prevention of epi-
ing to the allostatic load of the disease Bigal ME, Escandon R, Bronson M, Walter S, Sud-
sodic migraine, showed promising results
(Borsook et al., 2013). Given the enor- worth M, Huggins JP, Garzone P (2013)
(Bigal et al., 2013; Diener, 2014; Dodick et
mous burden to society (Ferrari, 1998), Safety and tolerability of LBR-101, a human-
al., 2014a,b; Russo, 2015). Remarkably, a there is an urgent imperative to focus on ized monoclonal antibody that blocks the
single injection may prevent or significantly better understanding the neurobiology of binding of CGRP to its receptor: results of the
reduce migraine attacks for 3 months. the disease to enable the discovery of phase 1 program. Cephalalgia 34:483– 492.
Given our growing understanding of CrossRef Medline
novel treatment approaches. Bigal ME, Walter S, Rapoport AM (2015) Ther-
the importance of prodromes (likely rep-
apeutic antibodies against CGRP or its recep-
resenting abnormal sensitivity to the fluc- References tor. Br J Clin Pharmacol. Advance online
tuation in hypothalamically regulated Aderjan D, Stankewitz A, May A (2010) Neuro-
publication. Retrieved April 2, 2015. doi:
nal mechanisms during repetitive trigemino-
homeostasis) and aura (likely represent- 10.1111/bcp.12591. CrossRef Medline
nociceptive stimulation in migraine patients.
ing the inherited cortical hyperexcitabil- Blau JN, Dexter SL (1981) The site of pain origin
Pain 151:97–103. CrossRef Medline
ity) in the pathophysiology of migraine, during migraine attacks. Cephalalgia 1:143–
Afridi SK, Giffin NJ, Kaube H, Friston KJ, Ward
147. CrossRef Medline
drugs that target ghrelin, leptin, and NS, Frackowiak RS, Goadsby PJ (2005) A
Bolay H, Reuter U, Dunn AK, Huang Z, Boas DA,
orexin receptors (Hirfanoglu et al., 2009; positron emission tomographic study in
Moskowitz MA (2002) Intrinsic brain activity
spontaneous migraine. Archives of Neurology
Peterlin et al., 2010; Sachdev and Mar- triggers trigeminal meningeal afferents in a mi-
62:1270 –1275. CrossRef Medline
mura, 2012; Chabi et al., 2015; Hoffmann Akerman S, Holland PR, Summ O, Lasalandra
graine model. Nat Med 8:136 –142. CrossRef
et al., 2015) may be considered for thera- MP, Goadsby PJ (2012) A translational in Medline
peutic development which is based on, vivo model of trigeminal autonomic cephalal- Borsook D, Burstein R (2012) The enigma of the
gias: therapeutic characterization. Brain 135: dorsolateral pons as a migraine generator.
their ability to restore proper hypotha- Cephalalgia 32:803– 812. CrossRef Medline
lamic control of stress, hyperphagia, adi- 3664 –3675. CrossRef Medline
Anttila V, Stefansson H, Kallela M, Todt U, Ter- Borsook D, May A, Goadsby P, Hargreaves R
posity, and sleep. All may be critical in windt GM, Calafato MS, Nyholt DR, Dimas (2012a) The migraine brain. New York:
reducing allostatic load and, conse- AS, Freilinger T, Müller-Myhsok B, Artto V, Oxford UP.
quently, in initiating the next migraine at- Inouye M, Alakurtti K, Kaunisto MA, Borsook D, Maleki N, Becerra L, McEwen B
Hämäläinen E, de Vries B, Stam AH, Weller (2012b) Understanding migraine through
tack (Borsook and Burstein, 2012;
CM, Heinze A, Heinze-Kuhn K, et al. (2010) the lens of maladaptive stress responses: a
Borsook et al., 2012b). model disease of allostatic load. Neuron 73:
Genome-wide association study of migraine
implicates a common susceptibility variant on 219 –234. CrossRef Medline
Brain modification 8q22.1. Nat Genet 42:869 – 873. CrossRef Borsook D, Aasted CM, Burstein R, Becerra L
Neuroimaging studies showing that brain Medline (2013) Migraine mistakes: error awareness.
Anttila V, Winsvold BS, Gormley P, Kurth T, Bet- Neuroscientist 20:291–304. CrossRef Medline
networks, brain morphology, and brain
tella F, McMahon G, Kallela M, Malik R, de Borsook D, Erpelding N, Lebel A, Linnman C,
chemistry are altered in episodic and Veggeberg R, Grant PE, Buettner C, Becerra L,
Vries B, Terwindt G, Medland SE, Todt U,
chronic migraineurs (Sprenger and Bor- McArdle WL, Quaye L, Koiranen M, Ikram Burstein R (2014) Sex and the migraine
sook, 2012; Charles, 2013; Denuelle and MA, Lehtimäki T, Stam AH, Ligthart L, We- brain. Neurobiol Dis 68:200 –214. CrossRef
Fabre, 2013) justify attempts to develop denoja J, et al. (2013) Genome-wide meta- Medline
therapies that widely modify brain networks analysis identifies new susceptibility loci for Burstein R, Jakubowski M (2004) Analgesic
migraine. Nat Genet 45:912–917. CrossRef triptan action in an animal model of intracra-
and their functions. Transcranial magnetic nial pain: a race against the development of
Medline
stimulation, which is thought to modify Apkarian AV (2008) Pain perception in relation central sensitization. Ann Neurol 55:27–36.
cortical hyperexcitability, is one such ap- to emotional learning. Curr Opin Neurobiol CrossRef Medline
proach (Schoenen et al., 2003; Stankewitz 18:464 – 468. CrossRef Medline Burstein R, Jakubowski M (2005) Unitary hy-
and May, 2008; Lipton and Pearlman, 2010; Apkarian AV, Sosa Y, Sonty S, Levy RM, Harden pothesis for multiple triggers of the pain and
RN, Parrish TB, Gitelman DR (2004) strain of migraine. J Comp Neurol 493:9 –14.
Sánchez-del-Río González, 2013). Another
Chronic back pain is associated with de- CrossRef Medline
approach for generalized brain modifica- creased prefrontal and thalamic gray matter Burstein R, Yamamura H, Malick A, Strassman AM
tion is cognitive behavioral therapy (Powers density. J Neurosci 24:10410 –10415. CrossRef (1998) Chemical stimulation of the intracranial
et al., 2013; Martin et al., 2014a,b). Medline dura induces enhanced responses to facial stim-
Burstein et al. • Migraine J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 • 6627

ulation in brain stem trigeminal neurons. J Neu- Dampney RA (2011) The hypothalamus and au- graine to society. Pharmacoeconomics 13:
rophysiol 79:964 –982. Medline tonomic regulation: an overview. In: Central 667– 676. CrossRef Medline
Burstein R, Yarnitsky D, Goor-Aryeh I, Ransil BJ, regulation of autonomic functions (Llewellyn- Ferrari MD, Klever RR, Terwindt GM, Ayata C,
Bajwa ZH (2000) An association between Smith IJ, Verberne AJM, eds), pp 47– 61. New van den Maagdenberg AM (2015) Migraine
migraine and cutaneous allodynia. Ann Neu- York: Oxford UP. pathophysiology: lessons from mouse models
rol 47:614 – 624. CrossRef Medline DaSilva AF, Granziera C, Snyder J, Hadjikhani N and human genetics. Lancet Neurol 14:65– 80.
Burstein R, Collins B, Jakubowski M (2004) De- (2007) Thickening in the somatosensory cor- CrossRef Medline
feating migraine pain with triptans: a race tex of patients with migraine. Neurology 69: Freilinger T, Anttila V, de Vries B, Malik R, Kallela
against the development of cutaneous allodynia. 1990 –1995. CrossRef Medline M, Terwindt GM, Pozo-Rosich P, Winsvold
Ann Neurol 55:19 –26. CrossRef Medline Davis KD, Dostrovsky JO (1988) Responses of B, Nyholt DR, van Oosterhout WP, Artto V,
Burstein R, Jakubowski M, Garcia-Nicas E, Kainz feline trigeminal spinal tract nucleus neurons Todt U, Hämäläinen E, Fernández-Morales J,
V, Bajwa Z, Hargreaves R, Becerra L, Borsook to stimulation of the middle meningeal artery Louter MA, Kaunisto MA, Schoenen J, Raita-
D (2010) Thalamic sensitization transforms and sagittal sinus. J Neurophysiol 59:648 – kari O, Lehtimäki T, Vila-Pueyo M, et al.
localized pain into widespread allodynia. Ann 666. Medline (2012) Genome-wide association analysis
Neurol 68:81–91. CrossRef Medline De Fusco M, Marconi R, Silvestri L, Atorino L, identifies susceptibility loci for migraine with-
Cao Y, Aurora SK, Nagesh V, Patel SC, Welch KM Rampoldi L, Morgante L, Ballabio A, Aridon out aura. Nat Genet 44:777–782. CrossRef
(2002) Functional MRI-BOLD of brainstem P, Casari G (2003) Haploinsufficiency of Medline
structures during visually triggered migraine. ATP1A2 encoding the Na⫹/K⫹ pump alpha2 Fuggetta G, Noh NA (2013) A neurophysiologi-
Neurology 59:72–78. CrossRef Medline subunit associated with familial hemiplegic cal insight into the potential link between
Chabi A, Zhang Y, Jackson S, Cady R, Lines C, migraine type 2. Nat Genet 33:192–196. transcranial magnetic stimulation, thalamo-
Herring WJ, Connor KM, Michelson D CrossRef Medline cortical dysrhythmia and neuropsychiatric
(2015) Randomized controlled trial of the Denuelle M, Fabre N (2013) Functional neuro- disorders. Exp Neurol 245:87–95. CrossRef
orexin receptor antagonist filorexant for mi- imaging of migraine. Rev Neurol (Paris) 169: Medline
graine prophylaxis. Cephalalgia 35:379 –388. 380 –389. CrossRef Medline Fuller-Thomson E, Schrumm M, Brennenstuhl S
CrossRef Medline Denuelle M, Fabre N, Payoux P, Chollet F, Geraud (2013) Migraine and despair: factors associated
Chang JC, Shook LL, Biag J, Nguyen EN, Toga G (2007) Hypothalamic activation in spon- with depression and suicidal ideation among Ca-
AW, Charles AC, Brennan KC (2010) Bi- taneous migraine attacks. Headache 47:1418 – nadian migraineurs in a population-based study.
phasic direct current shift, haemoglobin de- 1426. CrossRef Medline Depress Res Treat 2013:401487. CrossRef Medline
saturation and neurovascular uncoupling in Diamond S, Dalessio DJ (1982) The practicing Géraud G, Donnet A (2013) Migraine and hypo-
cortical spreading depression. Brain 133:996 – physicians approach to headaches, Ed 3. Bal-
thalamus. Rev Neurol (Paris) 169:372–379.
1012. CrossRef Medline timore, MD: Williams and Wilkins.
CrossRef Medline
Charles A (2013) Migraine: a brain state. Curr Dichgans M, Freilinger T, Eckstein G, Babini E,
Hadjikhani N (2008) Relevance of cortical
Opin Neurol 26:235–239. CrossRef Medline Lorenz-Depiereux B, Biskup S, Ferrari MD,
thickness in migraine sufferers. Expert Rev
Charles A, Brennan K (2009) Cortical spreading Herzog J, van den Maagdenberg AM, Pusch
Neurother 8:327–329. CrossRef Medline
depression-new insights and persistent ques- M, Strom TM (2005) Mutation in the neu-
Hadjikhani N, Sanchez Del Rio M, Wu O,
tions. Cephalalgia 29:1115–1124. CrossRef ronal voltage-gated sodium channel SCN1A
Schwartz D, Bakker D, Fischl B, Kwong KK,
Medline in familial hemiplegic migraine. Lancet 366:
Cutrer FM, Rosen BR, Tootell RB, Sorensen
Chasman DI, Schürks M, Anttila V, de Vries B, 371–377. CrossRef Medline
AG, Moskowitz MA (2001) Mechanisms of
Schminke U, Launer LJ, Terwindt GM, van Diener HC (2014) CGRP as a new target in pre-
migraine aura revealed by functional MRI in
den Maagdenberg AM, Fendrich K, Völzke H, vention and treatment of migraine. Lancet
human visual cortex. Proc Natl Acad Sci U S A
Ernst F, Griffiths LR, Buring JE, Kallela M, Neurol 13:1065–1067. CrossRef Medline
98:4687– 4692. CrossRef Medline
Freilinger T, Kubisch C, Ridker PM, Palotie A, Dodick DW, Goadsby PJ, Spierings EL, Scherer
Ferrari MD, Hoffmann W, et al. (2011) JC, Sweeney SP, Grayzel DS (2014a) Safety Hansen AJ, Zeuthen T (1981) Extracellular ion
Genome-wide association study reveals three and efficacy of LY2951742, a monoclonal an- concentrations during spreading depression
susceptibility loci for common migraine in the tibody to calcitonin gene-related peptide, for and ischemia in the rat brain cortex. Acta Physiol
general population. Nat Genet 43:695– 698. the prevention of migraine: a phase 2, ran- Scand 113:437– 445. CrossRef Medline
CrossRef Medline domised, double-blind, placebo-controlled Hansen JM, Lipton RB, Dodick DW, Silberstein
Coppola G, Schoenen J (2012) Cortical excit- study. Lancet Neurol 13:885– 892. CrossRef SD, Saper JR, Aurora SK, Goadsby PJ, Charles
ability in chronic migraine. Curr Pain Head- Medline A (2012) Migraine headache is present in the
ache Rep 16:93–100. CrossRef Medline Dodick DW, Goadsby PJ, Silberstein SD, Lipton aura phase: a prospective study. Neurology 79:
Coppola G, Pierelli F, Schoenen J (2007) Is the RB, Olesen J, Ashina M, Wilks K, Kudrow D, 2044 –2049. CrossRef Medline
cerebral cortex hyperexcitable or hyperre- Kroll R, Kohrman B, Bargar R, Hirman J, Hansen JM, Baca SM, Vanvalkenburgh P, Charles
sponsive in migraine? Cephalalgia 27:1427– Smith J (2014b) Safety and efficacy of A (2013) Distinctive anatomical and physio-
1439. CrossRef Medline ALD403, an antibody to calcitonin gene- logical features of migraine aura revealed by
Coppola G, Pierelli F, Schoenen J (2009) Habitua- related peptide, for the prevention of frequent 18 years of recording Brain 136:3589 –3595.
tion and migraine. Neurobiology of learning episodic migraine: a randomised, double- CrossRef Medline
and memory 92:249 –259. CrossRef Medline blind, placebo-controlled, exploratory phase Havanka-Kanniainen H, Tolonen U, Myllylä VV
Coppola G, Di Lorenzo C, Schoenen J, Pierelli F 2 trial. Lancet Neurol 13:1100 –1107. CrossRef (1988) Autonomic dysfunction in migraine:
(2013) Habituation and sensitization in pri- Medline a survey of 188 patients. Headache 28:465–
mary headaches. J Headache Pain 14:65. Ducros A, Denier C, Joutel A, Cecillon M, Lescoat 470. CrossRef Medline
CrossRef Medline C, Vahedi K, Darcel F, Vicaut E, Bousser MG, Headache Classification Committee of the Inter-
Cutrer FM, Olesen J (2006) Migraine with aura Tournier-Lasserve E (2001) The clinical national Headache Society (2013) The inter-
and their suforms. In: The headaches (Olesen spectrum of familial hemiplegic migraine as- national classification of headache disorders,
J, Goadsby PJ, Ramadan N, Tfelt-Hansen J, sociated with mutations in a neuronal calcium 3rd edition (beta version). Cephalalgia 33:
Welch KM, eds), pp 407– 421. Baltimore, MD: channel. N Engl J Med 345:17–24. CrossRef 629 – 808. CrossRef Medline
Lippincott, Williams and Wilkins. Medline Hirfanoglu T, Serdaroglu A, Gulbahar O, Cansu A
Cutrer FM, Sorensen AG, Weisskoff RM, Oster- Edvinsson L (2015) CGRP receptor antagonists (2009) Prophylactic drugs and cytokine and
gaard L, Sanchez del Rio M, Lee EJ, Rosen BR, and antibodies against CGRP and its receptor in leptin levels in children with migraine. Pediatr
Moskowitz MA (1998) Perfusion-weighted migraine treatment. Br J Clin Pharmacol. Ad- Neurol 41:281–287. CrossRef Medline
imaging defects during spontaneous migrain- vance online publication. Retrieved April 2, Hoffmann J, Supronsinchai W, Akerman S, An-
ous aura. Ann Neurol 43:25–31. CrossRef 2015. doi:10.1111/bcp.12618. CrossRef Medline dreou AP, Winrow CJ, Renger J, Hargreaves
Medline Ferrari MD (1998) The economic burden of mi- R, Goadsby PJ (2015) Evidence for orexiner-
6628 • J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 Burstein et al. • Migraine

gic mechanisms in migraine. Neurobiol Dis Lipton RB, Bigal ME, Ashina S, Burstein R, Silber- tical activity. Prog Neurobiol 39:337–388.
74:137–143. CrossRef Medline stein S, Reed ML, Serrano D, Stewart WF CrossRef Medline
Hosoya Y, Matsushita M, Sugiura Y (1983) A di- (2008) Cutaneous allodynia in the migraine McEwen BS (1998) Stress, adaptation, and dis-
rect hypothalamic projection to the superior population. Ann Neurol 63:148 –158. CrossRef ease. Allostasis and allostatic load. Ann N Y
salivatory nucleus neurons in the rat. A study Medline Acad Sci 840:33– 44. CrossRef Medline
using anterograde autoradiographic and ret- Liu Y, Broman J, Edvinsson L (2004) Central McEwen BS (2004) Protection and damage
rograde HRP methods. Brain Res 266:329 – projections of sensory innervation of the rat from acute and chronic stress: allostasis and
333. CrossRef Medline superior sagittal sinus. Neuroscience 129: allostatic overload and relevance to the patho-
Hosoya Y, Matsushita M, Sugiura Y (1984) Hy- 431– 437. CrossRef Medline physiology of psychiatric disorders. Ann N Y
pothalamic descending afferents to cells of or- Liveing E (1873) On megrim, sick headache. Ni- Acad Sci 1032:1–7. CrossRef Medline
igin of the greater petrosal nerve in the rat, as jmegen, The Netherlands: Arts and Boeve McEwen BS, Wingfield JC (2003) The concept
revealed by a combination of retrograde HRP Publishers. of allostasis in biology and biomedicine.
and anterograde autoradiographic tech- Llinás RR (1988) The intrinsic electrophysiolog- Horm Behav 43:2–15. CrossRef Medline
niques. Brain Res 290:141–145. CrossRef ical properties of mammalian neurons: in- Moskowitz MA (1993) Neurogenic inflamma-
Medline sights into central nervous system function. tion in the pathophysiology and treatment of
Kagan R, Kainz V, Burstein R, Noseda R (2013) Science 242:1654 –1664. CrossRef Medline migraine. Neurology 43:S16 –S20. Medline
Hypothalamic and basal ganglia projections Loewy AD (1990) Central autonomic path- Moskowitz MA, Macfarlane R (1993) Neuro-
to the posterior thalamus: possible role in ways. In: Central regulation of autonomic vascular and molecular mechanisms in mi-
modulation of migraine headache and photo- functions (Loewy AD, Spyer KM, eds). Ox- graine headaches. Cerebrovasc Brain Metab
phobia. Neuroscience 248:359 –368. CrossRef ford: Oxford UP. Rev 5:159 –177. Medline
Medline Maizels M, Scott B, Cohen W, Chen W (1996) Moulton EA, Burstein R, Tully S, Hargreaves R,
Karatas H, Erdener SE, Gursoy-Ozdemir Y, Lule Intranasal lidocaine for treatment of mi- Becerra L, Borsook D (2008) Interictal dys-
S, Eren-Koçak E, Sen ZD, Dalkara T (2013) graine: a randomized, double-blind, con- function of a brainstem descending modula-
Spreading depression triggers headache by ac- trolled trial. JAMA 276:319 –321. CrossRef tory center in migraine patients. PLoS One
tivating neuronal Panx1 channels. Science Medline 3:e3799. CrossRef Medline
339:1092–1095. CrossRef Medline Maleki N, Becerra L, Nutile L, Pendse G, Brawn J, Moulton EA, Becerra L, Maleki N, Pendse G, Tully
Kelman L (2004) The premonitory symptoms Bigal M, Burstein R, Borsook D (2011a) Mi- S, Hargreaves R, Burstein R, Borsook D
(prodrome): a tertiary care study of 893 mi- graine attacks the basal ganglia. Mol Pain 7:71. (2011) Painful heat reveals hyperexcitability
graineurs. Headache 44:865– 872. CrossRef CrossRef Medline of the temporal pole in interictal and ictal mi-
Medline graine states. Cereb Cortex 21:435– 448.
Maleki N, Becerra L, Nutile L, Pendse G, Brawn J,
Kelman L, Tanis D (2006) The relationship be- CrossRef Medline
Bigal M, Burstein R, Borsook D (2011b) Mi-
tween migraine pain and other associated Mutch WA, Hansen AJ (1984) Extracellular pH
graine attacks the basal ganglia. Mol Pain 7:71.
symptoms. Cephalalgia 26:548 –553. CrossRef changes during spreading depression and ce-
CrossRef
Medline rebral ischemia: mechanisms of brain pH reg-
Maleki N, Becerra L, Brawn J, Bigal M, Burstein R,
Kudrow L (1980) Cluster headache: mechanism ulation. J Cereb Blood Flow Metab 4:17–27.
Borsook D (2012a) Concurrent functional
and management. London: Oxford UP. CrossRef Medline
and structural cortical alterations in migraine.
Kudrow L, Kudrow DB, Sandweiss JH (1995) Noseda R, Burstein R (2013) Migraine pathophys-
Cephalalgia 32:607– 620. CrossRef Medline
Rapid and sustained relief of migraine attacks iology: anatomy of the trigeminovascular path-
Maleki N, Linnman C, Brawn J, Burstein R,
with intranasal lidocaine: preliminary find- way and associated neurological symptoms,
Becerra L, Borsook D (2012b) Her versus his
ings. Headache 35:79 – 82. CrossRef Medline cortical spreading depression, sensitization, and
migraine: multiple sex differences in brain
Larsson LI, Edvinsson L, Fahrenkrug J, Håkanson modulation of pain. Pain 154:S44 –S53 [Suppl
function and structure. Brain 135:2546 –2559.
R, Owman C, Schaffalitzky de Muckadell O, 1]. CrossRef Medline
Sundler F (1976) Immunohistochemical lo- CrossRef Medline Noseda R, Jakubowski M, Kainz V, Borsook D,
calization of a vasodilatory polypeptide (VIP) Maleki N, Becerra L, Brawn J, McEwen B, Bur- Burstein R (2011) Cortical projections of
in cerebrovascular nerves. Brain Res 113:400 – stein R, Borsook D (2013) Common hip- functionally identified thalamic trigemino-
404. CrossRef Medline pocampal structural and functional changes vascular neurons: implications for migraine
Lashley KS (1941) Patterns of cerebral integra- in migraine. Brain Struct Funct 218:903–912. headache and its associated symptoms. J Neu-
tion indicated by the scotomas of migraine. CrossRef Medline rosci 31:14204 –14217. CrossRef Medline
Arch Neurol Psychiatry 46:259 –264. Malick A, Strassman RM, Burstein R (2000) Tri- Noseda R, Kainz V, Borsook D, Burstein R
Leao A (1944) Spreading depression of activity geminohypothalamic and reticulohypotha- (2014) Neurochemical pathways that con-
in cerebral cortex. J Neurophysiol 7:359 –390. lamic tract neurons in the upper cervical verge on thalamic trigeminovascular neurons:
Leonardi M, Steiner TJ, Scher AT, Lipton RB spinal cord and caudal medulla of the rat. potential substrate for modulation of mi-
(2005) The global burden of migraine: mea- J Neurophysiol 84:2078 –2112. Medline graine by sleep, food intake, stress and anxiety.
suring disability in headache disorders with Maniyar FH, Sprenger T, Monteith T, Schankin PLoS One 9:e103929. CrossRef Medline
WHO’s classification of functioning, disabil- C, Goadsby PJ (2014) Brain activations in Nozaki K, Moskowitz MA, Maynard KI, Koketsu
ity and health (ICF). J Headache Pain 6:429 – the premonitory phase of nitroglycerin- N, Dawson TM, Bredt DS, Snyder SH (1993)
440. CrossRef Medline triggered migraine attacks. Brain 137:232– Possible origins and distribution of immuno-
Levy D, Jakubowski M, Burstein R (2004) Dis- 241. CrossRef Medline reactive nitric oxide synthase-containing
ruption of communication between periph- Martin PR, Reece J, Callan M, MacLeod C, Kaur nerve fibers in cerebral arteries. J Cereb Blood
eral and central trigeminovascular neurons A, Gregg K, Goadsby PJ (2014a) Behavioral Flow Metab 13:70 –79. CrossRef Medline
mediates the antimigraine action of 5HT management of the triggers of recurrent head- Obrenovitch TP, Urenjak J, Wang M (2002) Ni-
1B/1D receptor agonists. Proc Natl Acad Sci ache: a randomized controlled trial. Behav Res tric oxide formation during cortical spreading
U S A 101:4274 – 4279. CrossRef Medline Ther 61:1–11. CrossRef Medline depression is critical for rapid subsequent re-
Lipton RB, Pearlman SH (2010) Transcranial Martin PR, Mackenzie S, Bandarian-Balooch S, covery of ionic homeostasis. J Cereb Blood
magnetic simulation in the treatment of mi- Brunelli A, Broadley S, Reece J, Goadsby PJ Flow Metab 22:680 – 688. CrossRef Medline
graine. Neurotherapeutics 7:204 –212. CrossRef (2014b) Enhancing cognitive-behavioural ther- Ophoff RA, Terwindt GM, Vergouwe MN, van
Medline apyforrecurrentheadache:designofarandomised Eijk R, Oefner PJ, Hoffman SM, Lamerdin JE,
Lipton RB, Stewart WF, Diamond S, Diamond controlled trial. BMC Neurol 14:233. CrossRef Mohrenweiser HW, Bulman DE, Ferrari M,
ML, Reed M (2001) Prevalence and burden Medline Haan J, Lindhout D, van Ommen GJ, Hofker
of migraine in the United States: data from the McCormick DA (1992) Neurotransmitter ac- MH, Ferrari MD, Frants RR (1996) Familial
American Migraine Study II. Headache 41: tions in the thalamus and cerebral cortex and hemiplegic migraine and episodic ataxia
646 – 657. CrossRef Medline their role in neuromodulation of thalamocor- type-2 are caused by mutations in the Ca2⫹
Burstein et al. • Migraine J. Neurosci., April 29, 2015 • 35(17):6619 – 6629 • 6629

channel gene CACNL1A4. Cell 87:543–552. Schoonman GG, Evers DJ, Terwindt GM, van tive stimulation. Neurol Sci 32 [Suppl 1]:
CrossRef Medline Dijk JG, Ferrari MD (2006) The prevalence S165–S167. CrossRef Medline
Peterlin BL, Rapoport AM, Kurth T (2010) Mi- of premonitory symptoms in migraine: a Tucker DC, Saper CB (1985) Specificity of spi-
graine and obesity: epidemiology, mechanisms, questionnaire study in 461 patients. Cephalal- nal projections from hypothalamic and brain-
and implications. Headache 50:631– 648. gia 26:1209 –1213. CrossRef Medline stem areas which innervate sympathetic
CrossRef Medline Shechter A, Stewart WF, Silberstein SD, Lipton RB preganglionic neurons. Brain Res 360:159 –
Peters A, McEwen BS (2012) Introduction for (2002) Migraine and autonomic nervous system 164. CrossRef Medline
the allostatic load special issue. Physiol Behav function: a population-based, case-control study. Uddman R, Edvinsson L, Ekman R, Kingman T,
106:1– 4. CrossRef Medline Neurology 58:422–427. CrossRef Medline McCulloch J (1985) Innervation of the fe-
Powers SW, Kashikar-Zuck SM, Allen JR, LeCates Sherman SM (2005) Thalamic relays and corti- line cerebral vasculature by nerve fibers
SL, Slater SK, Zafar M, Kabbouche MA, cal functioning. Prog Brain Res 149:107–126. containing calcitonin gene-related peptide:
O’Brien HL, Shenk CE, Rausch JR, Hershey CrossRef Medline trigeminal origin and co-existence with
AD (2013) Cognitive behavioral therapy Sherman SM, Guillery RW (1998) On the ac- substance P. Neurosci Lett 62:131–136.
plus amitriptyline for chronic migraine in tions that one nerve cell can have on another: CrossRef Medline
children and adolescents: a randomized clini- distinguishing “drivers” from “modulators.” Valfrè W, Rainero I, Bergui M, Pinessi L (2008)
cal trial. JAMA 310:2622–2630. CrossRef Proc Natl Acad Sci U S A 95:7121–7126. Voxel-based morphometry reveals gray mat-
Medline CrossRef ter abnormalities in migraine. Headache 48:
Pozo-Rosich P, Oshinsky M (2005) Effects of di- Silberstein SD (1995) Migraine symptoms: re- 109 –117. CrossRef Medline
hydroergotamine (DHE) on central sensitiza- sults of a survey of self-reported migraineurs. Wahl M, Schilling L, Parsons AA, Kaumann A
tion of neurons in the trigeminal nucleus Headache 35:387–396. CrossRef Medline (1994) Involvement of calcitonin gene-
caudalis. Neurology 64:D131. Sluder G (1908) The role of the sphenopalatine related peptide (CGRP) and nitric oxide (NO)
Rodriguez-Raecke R, Niemeier A, Ihle K, Ruether ganglion in nasal headaches. N Y State J Med in the pial artery dilatation elicited by cortical
W, May A (2009) Brain gray matter decrease 27:8 –13. spreading depression. Brain Res 637:204 –210.
in chronic pain is the consequence and not the Sprenger T, Borsook D (2012) Migraine changes CrossRef Medline
cause of pain. J Neurosci 29:13746 –13750. Waldman SD (1993) Sphenopalatine ganglion
the brain: neuroimaging makes its mark. Curr
CrossRef Medline block— 80 years later. Reg Anesth 18:274 –
Opin Neurol 25:252–262. CrossRef Medline
Russell MB, Olesen J (1996) A nosographic anal- 276. Medline
Stankewitz A, May A (2008) Cortical dysbalance
ysis of the migraine aura in a general popula- Wang SJ, Fuh JL, Juang KD, Lu SR (2009) Mi-
in the brain in migraineurs— hyperexcitabil-
tion. Brain 119:355–361. CrossRef Medline graine and suicidal ideation in adolescents
ity as the result of sensitisation? Schmerz 22
Russo AF (2015) Calcitonin gene-related pep- aged 13 to 15 years. Neurology 72:1146 –1152.
[Suppl 1]:17–21. CrossRef Medline
tide (CGRP): a new target for migraine. Annu CrossRef Medline
Steriade M, Llinás RR (1988) The functional
Rev Pharmacol Toxicol 55:533–552. CrossRef Weiller C, May A, Limmroth V, Jüptner M, Kaube
states of the thalamus and the associated neu-
Medline H, Schayck RV, Coenen HH, Diener HC
ronal interplay. Physiol Rev 68:649 –742.
Sachdev A, Marmura MJ (2012) Metabolic syn- (1995) Brain stem activation in spontaneous
drome and migraine. Front Neurol 3:161. Medline human migraine attacks. Nat Med 1:658 – 660.
CrossRef Medline Steriade M, McCormick DA, Sejnowski TJ CrossRef Medline
Sánchez-del-Río González M (2013) Migraine: (1993) Thalamocortical oscillations in the Yarnitsky D, Goor-Aryeh I, Bajwa ZH, Ransil BI,
ignition of the brain. Rev Neurol 57:509 –514. sleeping and aroused brain. Science 262:679 – Cutrer FM, Sottile A, Burstein R (2003) 2003
Medline 685. CrossRef Medline Wolff Award: possible parasympathetic con-
Saper CB, Scammell TE, Lu J (2005) Hypotha- Stewart WF, Shechter A, Rasmussen BK (1994) Mi- tributions to peripheral and central sensitiza-
lamic regulation of sleep and circadian graine prevalence. A review of population-based tion during migraine. Headache 43:704 –714.
rhythms. Nature 437:1257–1263. CrossRef studies. Neurology 44:S17–S23. Medline CrossRef Medline
Medline Strassman AM, Raymond SA, Burstein R (1996) Zhang X, Levy D, Noseda R, Kainz V, Jakubowski
Saper CB, Fuller PM, Pedersen NP, Lu J, Scam- Sensitization of meningeal sensory neurons M, Burstein R (2010) Activation of menin-
mell TE (2010) Sleep state switching. Neu- and the origin of headaches. Nature 384:560 – geal nociceptors by cortical spreading depres-
ron 68:1023–1042. CrossRef Medline 564. CrossRef Medline sion: implications to migraine with aura.
Schock SC, Munyao N, Yakubchyk Y, Sabourin Sugaya E, Takato M, Noda Y (1975) Neuronal J Neurosci 30:8807– 8814. CrossRef Medline
LA, Hakim AM, Ventureyra EC, Thompson and glial activity during spreading depression Zhang X, Levy D, Kainz V, Noseda R, Jakubowski
CS (2007) Cortical spreading depression re- in cerebral cortex of cat. J Neurophysiol 38: M, Burstein R (2011) Activation of central
leases ATP into the extracellular space and pu- 822– 841. Medline trigeminovascular neurons by cortical spread-
rinergic receptor activation contributes to the Suzuki N, Hardebo JE (1993) The cerebrovascu- ing depression. Ann Neurol 69:855– 865.
induction of ischemic tolerance. Brain Res lar parasympathetic innervation. Cerebrovasc CrossRef Medline
1168:129 –138. CrossRef Medline Brain Metab Rev 5:33– 46. Medline Zhang X, Jakubowski M, Buettner C, Kainz V,
Schoenen J, Ambrosini A, Sándor PS, Maertens de Tessitore A, Russo A, Esposito F, Giordano A, Ta- Gold M, Burstein R (2013) Ezogabine
Noordhout A (2003) Evoked potentials and glialatela G, De Micco R, Cirillo M, Conte F, (KCNQ2/3 channel opener) prevents de-
transcranial magnetic stimulation in mi- d’Onofrio F, Cirillo S, Tedeschi G (2011) In- layed activation of meningeal nociceptors if
graine: published data and viewpoint on their terictal cortical reorganization in episodic mi- given before but not after the occurrence of
pathophysiologic significance. Clin Neuro- graine without aura: an event-related fMRI cortical spreading depression. Epilepsy Be-
physiol 114:955–972. CrossRef Medline study during parametric trigeminal nocicep- hav 28:243–248. CrossRef Medline

You might also like