Next Article in Journal
Application of Fluorescent Proteins for Functional Dissection of the Drosophila Visual System
Next Article in Special Issue
Testing the Role of Glutamate NMDA Receptors in Peripheral Trigeminal Nociception Implicated in Migraine Pain
Previous Article in Journal
Identification of the Ghrelin and Cannabinoid CB2 Receptor Heteromer Functionality and Marked Upregulation in Striatal Neurons from Offspring of Mice under a High-Fat Diet
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Neuroinflammation and Blood-Brain Barrier Permutability on Migraine

Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(16), 8929; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168929
Submission received: 22 July 2021 / Revised: 14 August 2021 / Accepted: 16 August 2021 / Published: 19 August 2021
(This article belongs to the Special Issue Molecular Research of Migraine: From Pathogenesis to Treatment)

Abstract

:
Currently, migraine is treated mainly by targeting calcitonin gene-related peptides, although the efficacy of this method is limited and new treatment strategies are desired. Neuroinflammation has been implicated in the pathogenesis of migraine. In patients with migraine, peripheral levels of pro-inflammatory cytokines, such as interleukin-1β (IL-1β) and tumor necrosis factor-α, are known to be increased. Additionally, animal models of headache have demonstrated that immunological responses associated with cytokines are involved in the pathogenesis of migraine. Furthermore, these inflammatory mediators might alter the function of tight junctions in brain vascular endothelial cells in animal models, but not in human patients. Based on clinical findings showing elevated IL-1β, and experimental findings involving IL-1β and both the peripheral trigeminal ganglion and central trigeminal vascular pathways, regulation of the Il-1β/IL-1 receptor type 1 axis might lead to new treatments for migraine. However, the integrity of the blood-brain barrier is not expected to be affected during attacks in patients with migraine.

1. Introduction

Migraine is now the sixth most common disease globally [1] and one of the three neurological disorders with the highest absolute burden of disability-adjusted life-years in the United States [2]. With a better understanding of the neurobiology of migraine and the 5-hydroxytryptamine (5-HT) receptor, or serotonin receptor, the burden on patients with migraine has been lightened with the creation of selective 5-HT1B/1D agonists called triptans [3]. However, migraine still imposes a significant burden on individuals and society from childhood to adulthood. An important issue in the study of migraine is the identification of neurotransmitters and neuromodulators that may be involved in the pathophysiology of migraine [4].
Currently, the dominant role of the calcitonin gene-related peptide (CGRP) pathway is the main focus of migraine research and has been validated and clinically applied by a series of translational studies [5]. An overview of the pathophysiology of migraine is shown in Figure 1, with an emphasis on the role of CGRP in the trigemino-vascular system. However, although CGRP has also been implicated in the activation of inflammatory cells, there is a limit to the effectiveness of CGRP-targeted therapy [6]. As novel treatment strategies are being explored, the concept of neuroinflammation could be a productive option to investigate.
Cortical spreading depression (CSD), relevant to the development of migraine aura, has been shown to activate the trigemino–vascular system (TVS). Anti-temporal conduction of trigeminal ganglion neurons leads to the release of neuropeptides, including calcitonin gene-related peptide (CGRP), from their nerve terminals, leading to vasodilation and plasma extravasation. The dura is densely innervated by sensory nerve fibers that contain these neuropeptides originating from the trigeminal ganglion (TG), causing neurogenic inflammation in the dura (A). In contrast, direct conduction of TG neurons generates a pain sensation via the activation of c-fos in trigeminal nucleus caudalis (TNC) (B), which is eventually perceived as a headache. CGRP receptors are observed in TGs and TNCs but not in peripheral trigeminal nerve terminals, suggesting that CGRP receptor antagonists may suppress neurons in TGs and TNCs. An explanation of how CSD stimulates TVS is given in Figure 1.
Neurogenic inflammation in migraine is mainly characterized by the release of neuropeptides such as CGRP and substance P from the trigeminal nerve, leading to arterial vasodilation, plasma protein extravasation, and mast cell degranulation. The involvement of these neuropeptides in migraine is evident [7,8], and pro-inflammatory cytokines or chemokines may be involved in this series of reactions [9,10].
Neuroinflammation is essentially defined as an inflammatory response in the brain and spinal cord, which is modulated by the production of cytokines, chemokines, reactive oxygen species (ROS), and secondary messengers such as nitric oxide (NO) and prostaglandins [11,12]. They are mainly produced by activated microglia and astrocytes [11,12], and neurovascular units comprising neurons, pericytes, and endothelial cells [13,14]. Neuroinflammation is closely related to central nervous system (CNS) diseases, such as multiple sclerosis and epilepsy, and the regulation of cytokines has revealed new therapeutic aspects.
This review explores the current literature on neuroinflammation. It targets neuroinflammation from the perspective of cytokines in the pathogenesis of migraine and aims to identify new directions for research on therapeutic interventions for migraine targeting neuroinflammation.

2. Summary of Search

This narrative literature review is based on studies on the involvement of oxidative stress, neuroinflammation, and mitochondrial dysfunction in migraine, and the effectiveness of riboflavin as a prophylactic treatment for migraine. A literature search was conducted in the PubMed database and included articles published up to June 2021. The keywords used in the search were “migraine” and “inflammation” or “neuroinflammation” or “cytokine” or “chemokine” or “blood brain barrier.”

3. Clinical Evidence of Neuroinflammation

There have been a number of studies on cytokines and chemokines in patients with migraine, including studies performed during the attack, in the interictal period, and even during the post-attack period.
During migraine attacks (ictal period), interleukin-1β (IL-1β), IL-6, IL-8, and tumor necrosis factor-α (TNF-α) are increased [15,16,17,18,19,20]. Pediatric patients without aura showed higher levels of IL-1β than those with aura [17]. However, some reports have shown no significant difference in TNF-α and IL-1β levels between attacks and attack-free periods [21,22]. Decreased levels of the anti-inflammatory cytokines IL-4 and IL-5 were reported during the attacks [18,23]. Increased levels of IL-10 (an anti-inflammatory cytokine) during attacks, compared with the interictal period [16,24,25], and a decrease in IL-10 by sumatriptan (one of the triptans) were found [25].
During headache-free periods (interictal period), increased pro-inflammatory levels of IL-1β, IL-6, TNF-α, and chemokines such as IL-8, macrophage inflammatory protein-1α (MIP-1α), and C–C chemokine ligand 5 (CCL5) were observed in patients [24,26,27,28,29]. Conversely, IL-10 levels were found to be similar [15] or decreased [25,26,27] when compared with healthy individuals.
Studies measuring interictal cytokine concentrations in patients with migraine have reported conflicting results, with some showing no differences between migraine and control groups for TNF-α and soluble TNF receptors TNF-R1 and -R2 [22,30], while others show increased levels in patients with migraine [26]. IL-6 and IL-8 levels were lower than those in the control group [28].
Some reports have examined the association between these markers, headache severity, and comorbidity. Plasma TNF-α is negatively correlated with anxiety scores [28], and the serum MIP-1 or MIP-1:CD14 ratio was reported to be associated with the pain index and the severity of headache [31]. However, elevated serum IL-8 and MIP-1α do not appear to be associated with comorbid psychiatric disorders or allodynia [29].

3.1. Serial Analysis of Cytokines

While there is some directionality, such as an increase in pro-inflammatory cytokines during both migraine attacks and interictal periods, no definite consensus has been reached. The duration of headache attacks varies widely, which may explain the inconsistent results in terms of sampling timing. Serial analysis of cytokines from jugular venous blood by Sarchielli et al. yielded consistent and robust data which, quite intriguingly, were completely different from data taken from the periphery [32,33]. Significant increases in TNF-α, IL-6, nuclear factor (NF)-κB, and soluble intercellular adhesion molecule-1 (sICAM-1) were observed in parallel within 2 h of attack onset compared with the time of catheter insertion [33]. CGRP increased significantly after 1 h and IL-8 reached its highest level at 4 h, while the other two chemokines, RANTES (CCL5) and monocyte chemoattractant protein-1 (MCP-1), did not significantly change at any time point [32]. While a slight increase in IL-1β was observed from 1 to 4 h, the levels of IL-1β decreased, reaching values at the end of the attack [18]. The levels of cytokines, selectins, vascular adhesion molecules, and adhesion molecules in the peripheral blood of patients with migraine were found to be unaltered at each time point of the study [32,33]. The results of these investigations are consistent and significant over time, illustrating the difficulty of examining cytokines in patients with migraine. Carotid artery blood may provide clearer information than peripheral blood regarding biochemical and neurotransmitter changes in the cerebral circulation, and may be more characteristic of migraine attacks.

3.2. Cerebral Spinal Fluid (CSF)

Despite the normal serum TNF-α levels in patients with chronic migraine (CM), elevated TNF-α levels in the CSF have been identified [34]. Although IL-1 receptor antagonist (IL-1RA), MCP-1, and transforming growth factor-β1 were significantly increased in the CSF of patients with episodic tension-type headache and in migraine patients with and without aura compared to those without pain, the increases were not sufficiently different between these headache types [35]. A comparison of CSF and plasma or serum cytokine levels would provide some insight into the origin of cytokines, although the assessment of CSF cytokines in patients with migraine is limited. Due to ethical issues, it may be difficult to examine CSF in patients with migraine for the foreseeable future, hence the value of studying available reports.

3.3. Genetic Analysis

Consistent with the predominant theory of migraine that emphasizes vascular etiology, a genome-wide association showed that loci associated with migraine are rich in single nucleotide polymorphisms in vascular and smooth muscle tissues [36]. However, the frequency of the TNF-α-308 GG genotype was lower in the patient group than in the control group [37], suggesting a link between migraine and inflammation. In addition, current genetic analysis of muscle biopsies in the calvarial periosteum from patients with CM (where the pain was situated) found the expression of pro-inflammatory genes (e.g., CCL8 and toll-like receptor 2) to be significantly increased in patients with CM, attesting to muscle tenderness, whereas the expression of genes that suppress inflammation and immune cell differentiation (e.g., IL-10 receptor subunit alpha and colony-stimulating factor 1 receptor) was decreased [38]. These results might reflect the activation of the neuroinflammatory cascade in migraine pathogenesis.

4. Experimental Evidence of Neuroinflammation

To investigate the relationship between migraine and neuroinflammation, two main models, the cortical spreading depolarization (CSD) mouse model and the nitroglycerin (NTG)-induced rat model, were used, and the findings from each model are as follows.

4.1. Inflammation Response Provoked by CSD

CSD is recognized as a characteristic feature of migraine with aura, representing a strong wave of neuronal depolarization with glial and vascular activation [39,40], which was originally published by Leao [41]. The clinical significance of CSD, which can occur after ischemic, hemorrhagic, or traumatic brain injury, has been emphasized and recognized [42,43]. In experimental animals, CSD can activate trigeminal nociceptors [44] and is inhibited by drugs used for migraine prophylaxis [45]. The mouse model of CSD was used as an experimental model relevant to the pathophysiology of migraine.
CSD can trigger a substantial inflammatory response via the opening of neuronal Pannexin 1 megachannels and activation of caspase-1, followed by the release of high-mobility group box 1 from neurons and the activation of astrocytes [46]. Furthermore, the development of CSD has been shown to cause inflammation of the meninges by activating macrophages and mast cells and enhancing the production of several inflammatory cytokines, such as IL-1β, IL-6, and TNF-α [47,48,49,50,51].
In vitro analysis of spreading depression in hippocampal organotypic cultures [52] and astrocytes [51] reported upregulation of pro-inflammatory cytokines, such as IL-6, IL-1β, and TNF-α. Additionally, increased levels of IL-1β, IL-6, and TNF-α were detected in the representative studies of in vivo rodent models [49,53,54]. However, some studies could not detect similar findings [55,56].
These conflicting and inconsistent results might be due to the invasive method of CSD induction, which might induce an inflammatory response [55]. In transgenic mice expressing channelrhodopsin-2 in neurons (Thy1-ChR2 YFP transgenic mice) [57,58], optogenetics enabled an almost non-invasive induction of CSD [59,60]. Takizawa et al. demonstrated that non-invasive induction of CSD also evoked a potent pro-inflammatory response in mice, such as cytokines IL-1β, TNF-α, IL-6, chemokine CCL2, and cell adhesion molecules (ICAM-1 and vascular cell adhesion molecule-1) after a single cell or multiple CSDs [60]. Interestingly, these responses were attenuated in IL-1 receptor type 1 (IL-1R1) knockout mice, suggesting the involvement of IL-1β as an upstream mediator [60]. However, the inflammatory response induced by CSD was suppressed by dexamethasone but not by ibuprofen, the most commonly used acute migraine medication [60]. Ibuprofen, an anti-inflammatory agent, might act downstream after an inflammatory response, although a discrepancy between clinical practice and basic research appears to exist. The efficacy of dexamethasone in acute migraine is reported to have a negative effect on improvement [61] and short-term prevention [62,63] of migraine headaches. The suppression of the inflammatory response by dexamethasone is a recognized fact, and there is limited evidence of clinical findings with dexamethasone. There is no doubt that there is concrete evidence of neuroinflammation caused by CSD, and the suppression of inflammation by CSD might not necessarily translate into migraine treatment.

4.2. Spontaneous Migraine-like Mouse Model Using NTG

NTG administration can induce spontaneous migraine-like headaches in rodents. NTG is a vasodilator that acts as a nitric oxide donor and reproduces multiple symptoms of migraine, inducing hyperalgesia at the trigeminal and spinal levels [64].
In a rat model of meningeal inflammation following NTG, pro-inflammatory cytokine IL-1β increased in the dura mater and IL-6 increased in dural macrophages and the CSF, while NOS mRNA up-regulation was observed within the first hours after the infusion [65,66]. The increased transcriptional activity of inflammatory cytokines (IL-6 and TNF-α), CGRP, and nNOS with NTG-induced hyperalgesia was also detected in the trigeminal ganglia, cervical spinal cord, and medulla-pons, and blocked by the injection of kynurenic acid analog 1, an endogenous regulator of glutamate activity [67].
In addition, the peripheral inhibition of fatty acid amide hydrolase, an enzyme that deactivates the endocannabinoid anandamide, can lead to an antihyperalgesic effect with a decrease in CGRP, substance P, and TNF-α and IL-6 mRNA in two complementary rat models of episodic CM [68].
In the central area of the trigeminovascular pathway, activation of the microglial leucine-rich repeat pyrin containing protein-3 (NLRP3) inflammasome mediates the release of IL-1β and contributes to central sensitization in the NTG-induced migraine model. Additionally, NTG-induced CM-like pain was paralleled by activation of the NLRP3 inflammasome [69]. High levels of extracellular K+ ions, mitochondrial dysfunction, and the production of ROS are also known to activate NLRP3, which has been shown to underlie the development of migraine [70,71,72,73]. MCC950, an inhibitor of NLRP3 inflammasome, might also have a preventive effect on migraine occurrence.

5. Presence of Blood-Brain Barrier (BBB) Permutability in Migraine

The presence or absence of BBB impairment associated with neuroinflammation is important when considering therapeutic strategies. Studying a drug’s permeability from the periphery to the brain, and its potential to infiltrate cells such as monocytes that introduce cytokines into the brain, will lead to new therapeutic strategies in the future.

5.1. Experimental Research of BBB Permutability

Experimental studies of rat models resembling primary headache have shown that CSD modulates the permeability of the BBB by activating matrix metalloproteinase-9 (MMP-9) from 3 h after CSD induction, peaking at 24 h [74]. A recent study in awake rats also found that cortical BBB leakage began 0.5 h after CSD induction and resolved within 6 h, without altering the tight junction proteins occludin or claudin-5 [75]. CSD-induced BBB opening to water and large molecules is mediated by increased endothelial transcytosis. These phenomena are dependent on caveolin-1 and rho-kinase 2; in contrast, endothelial tight junctions, pericytes, and basement membranes are maintained after CSD [76]. This change in BBB permeability is not destructive and may be a transient alteration associated with an inflammatory response. It may also directly affect the access of agents to centrally located targets during migraine attacks [77]. Indeed, increased uptake of sumatriptan into the brain was detected, in association with transient BBB permeability, in a KCl-induced model of episodic headache [75]. Previous reports suggest that dural inflammation induced by trigeminal ganglion (TG) stimulation does not affect the integrity of the BBB [78].

5.2. Clinical Studies of BBB Permutability

Previous clinical studies have found no association between primary headache and BBB opening [79]. Elevations of MMP-9 [80] and ICAM-1 [19], which are considered typical markers suggestive of BBB disorders, are observed in patients with migraine. An MMP-9 haplotype was reported to affect circulating MMP-9 levels in women with migraine [81].
One study reported a significant elevation of endothelial cell-specific molecule-1 (ESM-1) and claudin-5 in the migraine attack group [20]. ESM-1 is expressed in the vascular endothelium and is regulated by a number of cytokines, including IL-1β and TNF-α, and growth factors. Claudin-5 regulates BBB permeability and is important in sustaining the integrity of cerebrovascular endothelial cells [82,83]. A positive correlation was also found between Visual Analog Scale scores and the levels of ESM-1 and claudin-5 [20]. In addition to the association of BBB disorders with the pathogenesis of migraine, the results suggest a link between the clinical severity of migraine and BBB disorders.
However, several recent magnetic resonance imaging (MRI) studies also demonstrated no increased BBB permeability during spontaneous migraine attacks with [84] and without aura [85]. These studies were performed relatively early, between 6.5 and 7.6 h from attack onset to MRI scanning [84,85], with reference to rodent studies [74]. In addition, glyceryl trinitrate-induced dural inflammation could not affect BBB function during ictal and interictal periods, and the passage of dihydroergotamine through the BBB was also negative [86]. These clinical results indicate that the integrity of the BBB is unlikely to be affected during a migraine attack, contrary to the results from animal models.

6. Participation of Pericytes in Migraine

Pericytes are a neurovascular unit component of the BBB and play an important role in the integrity of the BBB. Pericyte degeneration and/or dysfunction contribute to the loss of BBB integrity, which is an early hallmark of several neurodegenerative and inflammatory conditions [13,87,88].
Capillary pericytes are also shown to play an active role in the regulation of cortical vasculature during and after CSD [89]. In fact, prolonged vasoconstriction caused by CSD was revealed to be strongest in primary capillaries, where pericytes have a sustained increase in calcium levels. Somatosensory stimulation after CSD causes no further changes in the diameter of capillaries or calcium in pericytes, suggesting that pericytes play a critical role in long-term oligemia after CSD [89]. Current research has demonstrated that brain pericytes respond to inflammatory signals, including IL-1β and TNF-α [87,90,91,92,93]. Pericytes may act as sensors for the inflammatory response in the CNS [92,94]. Based on BBB integrity and systemic peripheral inflammation, pericytes are speculated to play a pivotal role in the pathogenesis of epilepsy during neuroinflammation [14]. Structural changes in pericytes associated with epileptic seizures are exacerbated by IL-1β rather than TNF-α [95]. It is uncertain whether migraine, like epilepsy, is associated with BBB disorder and neuroinflammation, and experimental results targeting pericytes have not been obtained. However, structural changes in pericytes and the endothelial cells of microvessels were also observed in patients with familial hemiplegic migraine (FHM) [96]. It is known that both migraine and epilepsy share pathologies and clinical features to a certain extent, through similar underlying pathophysiological mechanisms, and antiepileptics such as valproic acid or topiramate are effective in treating patients with both disorders [97,98]. Therefore, the prevention or treatment of pericyte constriction may become a therapeutic target in migraine [89,99].

7. Prospect of Therapeutic Benefit via the IL-1β/IL-1R1 Axis

IL-1β is a master cytokine that drives both brain and systemic inflammation, which activates a ubiquitous cell surface receptor, IL-1R1. The IL-1β/IL-1R1 axis activates a cascade of inflammatory molecules, including other cytokines and chemokines. In vivo analysis of meningeal nociceptors in the TG demonstrated that IL-1β activates and sensitizes meningeal nociceptors [100] and has been proposed as a key mediator in trigeminal activation after CSD [46]. In addition to the experimental models described above, increased gene expressions of IL-1β and IL-1RA were also detected in a mouse model of FHM type 1 (FHM-1) [101,102]. IL-1β mediates the pro-inflammatory process, promotes the activation of trigeminal satellite cells, and supports the cross-excitation of satellite glial cells and neurons in the TG [11,103].
IL-1 also plays an important role in the pathogenesis of epilepsy, and its antagonist, IL-1RA, exhibits brain-protective effects as well as potent anticonvulsant effects [104,105]. Anakinra, a human recombinant version of IL-1RA, has emerged as a new treatment option for intractable epilepsy and exerts its neuroprotective effects by suppressing epileptic seizures in fairly intractable epilepsy [106,107,108].
The usefulness of anakinra was also suggested for CNS diseases such as stroke [109] and subarachnoid hemorrhage [110]. Interestingly, anakinra can improve headache as well with the resolution of CNS inflammation in patients with cryopyrin-associated periodic fever syndromes—a spectrum of rare inherited autoinflammatory syndromes [111,112]. Based on these facts, we should also expect anakinra to be effective in treating migraine. However, in conditions such as migraine where the BBB is not damaged or is only compromised to a limited extent, unlike epilepsy, there is some concern that anakinra may have difficulty reaching the brain [113,114]. Anakinra was found to cross the BBB in a dose-dependent manner in nonhuman primates [115]; in fact, a dose-dependent effect was observed in patients with epilepsy [108].
The inhibitory effect of anakinra on neuroinflammation has been reported not only in the CNS but also in the peripheral nervous system. In mouse models of familial amyloid polyneuropathy, anakinra inhibits transthyretin expression in Schwann cells [116] and extracellular deposition, and protects against unmyelinated fibrillary degeneration [117].
In experimental models, along the course of the IL-1β/IL-1R1 axis, inhibition of the NLRP3 inflammasome by MCC950, but not by anakinra, led to suppressed NTG-induced hyperalgesia and decreased protein expression levels of c-fos and CGRP in the trigeminal nucleus caudalis. MCC950 is the most advanced NLRP3 small molecule inhibitor [118], and there might be an option other than anakinra for IL-1β suppression through NLRP3 inflammasome inhibition by MCC950 [69] (Figure 2).
Recently, curcumin, a dietary polyphenol, has been demonstrated to significantly inhibit IL-1β and TNF-α expression, oxidative stress, and protein accumulation, and to eventually protect neurons from neurodegeneration in several different disease models [119,120,121,122]. Additionally, curcumin has been shown to have a preemptive analgesic and antioxidant effect on experimental migraine [123].
CSD can trigger Pannexin-1 megachannel opening and mediate the formation of the inflammasome, comprising apoptosis-associated speck-like protein (ASC), NLRP3, and pro-caspase-1, in neurons of the cortex area. Pro-caspase-1, released by the formation of inflammasomes, is activated to cleave and mature pro-IL-1β. Additionally, IL-1β induces a substantial neuroinflammatory signal that alerts adjacent cells and eventually reaches the trigeminal nervous system around the meningeal vessels, activating this nervous system and resulting in a headache. In the CNS, CSD can induce the production of IL-1β by neurons, astrocytes, and microglia. IL-1β binds to the type I IL-1 receptor and activates the transcription factor NF-κB, which promotes the production of pro-inflammatory cytokines and generates an inflammatory cascade. Anakinra antagonizes the effects of IL-1β by inhibiting IL-1R1, and MCC950 stimulates the blockade of these neuroinflammatory cascades by inhibiting NLRP3, which might suppress the headache.

8. Conclusions

In this review, we present evidence for the substantive role of neuroinflammation in migraine. There is increasing evidence of abnormal cytokine production in both human and experimental models, and the inhibition of neuroinflammation may contribute to the improvement of migraine pathology [67,68]. Based on the described reviews, the Il-1β/IL-1R1 axis in particular plays a crucial role in both the peripheral TG [100,103,124] and trigeminovascular pathway in the CNS [69]. Thus, regulation of the IL-1β cascade by anakinra and MCC950 may lead to new therapeutic options for migraine.
Although these inflammatory mediators, including IL-1β, have been known to lead to BBB impairment, at present, BBB disturbances are indicated only in rodent models, while human studies have been negative. An elevation of inflammatory mediators, including MMP-9, and structural changes in pericytes, were detected in patients with migraine. Disruption of the BBB could not be completely ruled out, and stabilization of the BBB might be a potential therapeutic target for migraine in the future.

Author Contributions

Conceptualization, G.Y.; Investigation, S.S. (Shinji Suzuki), N.M., M.T., K.K., T.T. and S.S. (Shunsuke Suzuki); resources, S.M., Y.W., Y.I., S.G. and S.O.; Writing—original draft preparation, G.Y.; Writing—review and editing, Y.K.; Supervision, H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

We confirm that we have read the journal’s position on the issues associated with ethical publication and affirm that this report is consistent with these guidelines.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets generated and/or analyzed during the current study are available at the PubMed database repository (https://pubmed.ncbi.nlm.nih.gov/ (access date: 1 June 2021)).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BBBblood-brain barrier
ILinterleukin
MMP-9metalloproteinase-9
MRImagnetic resonance image
TNF-αtumor necrosis factor-α
ESM-1endothelial cell-specific molecule-1
TGtrigeminal ganglia
ROSreactive oxygen species
NOnitric oxide
CGRPcalcitonin gene-related peptide
CNScentral nervous system
ICAM-1intercellular adhesion molecule-1
CSFcerebral spinal fluid
CSDcortical spreading depolarization
NTGnitroglycerin
NLRP3leucine-rich repeat pyrin containing protein-3
5-HT5-hydroxytryptamine
MIP-1αmacrophage inflammatory protein-1α
CMchronic migraine
MCP-1monocyte chemoattractant protein-1
CCL5C–C chemokine ligand 5
IL-1R1IL-1 receptor type 1

References

  1. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017, 390, 1211–1259. [CrossRef] [Green Version]
  2. Feigin, V.L.; Vos, T.; Alahdab, F.; Amit, A.M.L.; Bärnighausen, T.W.; Beghi, E.; Beheshti, M.; Chavan, P.P.; Criqui, M.H.; Desai, R.; et al. Burden of Neurological Disorders Across the US From 1990–2017: A Global Burden of Disease Study. JAMA Neurol. 2021, 78, 165–176. [Google Scholar] [CrossRef] [PubMed]
  3. Humphrey, P.P.; Feniuk, W.; Perren, M.J.; Beresford, I.J.; Skingle, M.; Whalley, E.T. Serotonin and migraine. Ann. N. Y. Acad. Sci. 1990, 600, 587–598; discussion 598–600. [Google Scholar] [CrossRef] [PubMed]
  4. Edvinsson, L.; Uddman, R. Neurobiology in primary headaches. Brain Res. Brain Res. Rev. 2005, 48, 438–456. [Google Scholar] [CrossRef] [Green Version]
  5. Edvinsson, L.; Haanes, K.A.; Warfvinge, K.; Krause, D.N. CGRP as the target of new migraine therapies—successful translation from bench to clinic. Nat. Rev. Neurol. 2018, 14, 338–350. [Google Scholar] [CrossRef]
  6. Raddant, A.C.; Russo, A.F. Calcitonin gene-related peptide in migraine: Intersection of peripheral inflammation and central modulation. Expert Rev. Mol. Med. 2011, 13, e36. [Google Scholar] [CrossRef] [Green Version]
  7. Ramachandran, R. Neurogenic inflammation and its role in migraine. Semin. Immunopathol. 2018, 40, 301–314. [Google Scholar] [CrossRef]
  8. Malhotra, R. Understanding migraine: Potential role of neurogenic inflammation. Ann. Indian Acad. Neurol. 2016, 19, 175–182. [Google Scholar] [CrossRef]
  9. Gao, Y.J.; Ji, R.R. Chemokines, neuronal-glial interactions, and central processing of neuropathic pain. Pharmacol. Ther. 2010, 126, 56–68. [Google Scholar] [CrossRef] [Green Version]
  10. Edvinsson, L.; Haanes, K.A.; Warfvinge, K. Does inflammation have a role in migraine? Nat. Rev. Neurol. 2019, 15, 483–490. [Google Scholar] [CrossRef]
  11. DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139 (Suppl. 2), 136–153. [Google Scholar] [CrossRef] [Green Version]
  12. Norden, D.M.; Trojanowski, P.J.; Villanueva, E.; Navarro, E.; Godbout, J.P. Sequential activation of microglia and astrocyte cytokine expression precedes increased Iba-1 or GFAP immunoreactivity following systemic immune challenge. Glia 2016, 64, 300–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Armulik, A.; Genové, G.; Betsholtz, C. Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises. Dev. Cell 2011, 21, 193–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Yamanaka, G.; Takata, F.; Kataoka, Y.; Kanou, K.; Morichi, S.; Dohgu, S.; Kawashima, H. The Neuroinflammatory Role of Pericytes in Epilepsy. Biomedicines 2021, 9, 759. [Google Scholar] [CrossRef] [PubMed]
  15. Covelli, V.; Munno, I.; Pellegrino, N.M.; Di Venere, A.; Jirillo, E.; Buscaino, G.A. Exaggerated spontaneous release of tumor necrosis factor-alpha/cachectin in patients with migraine without aura. Acta Neurol. 1990, 12, 257–263. [Google Scholar]
  16. Perini, F.; D’Andrea, G.; Galloni, E.; Pignatelli, F.; Billo, G.; Alba, S.; Bussone, G.; Toso, V. Plasma Cytokine Levels in Migraineurs and Controls. Headache J. Head Face Pain 2005, 45, 926–931. [Google Scholar] [CrossRef]
  17. Kacinski, M.; Gergont, A.; Kubik, A.; Steczkowska-Klucznik, M. Proinflammatory cytokines in children with migraine with or without aura. Przegl. Lek. 2005, 62, 1276–1280. [Google Scholar]
  18. Sarchielli, P.; Alberti, A.; Baldi, A.; Coppola, F.; Rossi, C.; Pierguidi, L.; Floridi, A.; Calabresi, P. Proinflammatory cytokines, adhesion molecules, and lymphocyte integrin expression in the internal jugular blood of migraine patients without aura assessed ictally. Headache 2006, 46, 200–207. [Google Scholar] [CrossRef]
  19. Wang, F.; He, Q.; Ren, Z.; Li, F.; Chen, W.; Lin, X.; Zhang, H.; Tai, G. Association of serum levels of intercellular adhesion molecule-1 and interleukin-6 with migraine. Neurol. Sci. 2015, 36, 535–540. [Google Scholar] [CrossRef] [PubMed]
  20. Yücel, M.; Kotan, D.; Gurol Çiftçi, G.; Çiftçi, I.H.; Cikriklar, H.I. Serum levels of endocan, claudin-5 and cytokines in migraine. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 930–936. [Google Scholar]
  21. Van Hilten, J.J.; Ferrari, M.D.; Van der Meer, J.W.M.; Gijsman, H.J.; Looij, B.J., Jr. Plasma interleukin-1, tumour necrosis factor and hypothalamic-pituitary-adrenal axis responses during migraine attacks. Cephalalgia 1991, 11, 65–67. [Google Scholar] [CrossRef] [PubMed]
  22. Tanure, M.T.; Gomez, R.S.; Hurtado, R.C.; Teixeira, A.L.; Domingues, R.B. Increased serum levels of brain-derived neurotropic factor during migraine attacks: A pilot study. J. Headache Pain 2010, 11, 427–430. [Google Scholar] [CrossRef] [Green Version]
  23. Martelletti, P.; Stirparo, G.; Morrone, S.; Rinaldi, C.; Giacovazzo, M. Inhibition of intercellular adhesion molecule-1 (ICAM-1), soluble ICAM-1 and interleukin-4 by nitric oxide expression in migraine patients. J. Mol. Med. 1997, 75, 448–453. [Google Scholar] [CrossRef] [PubMed]
  24. Fidan, I.; Yüksel, S.; Ymir, T.; Irkeç, C.; Aksakal, F.N. The importance of cytokines, chemokines and nitric oxide in pathophysiology of migraine. J. Neuroimmunol. 2006, 171, 184–188. [Google Scholar] [CrossRef] [PubMed]
  25. Munno, I.; Marinaro, M.; Bassi, A.; Cassiano, M.A.; Causarano, V.; Centonze, V. Immunological aspects in migraine: Increase of IL-10 plasma levels during attack. Headache 2001, 41, 764–767. [Google Scholar] [CrossRef]
  26. Boćkowski, L.; Sobaniec, W.; Zelazowska-Rutkowska, B. Proinflammatory plasma cytokines in children with migraine. Pediatr. Neurol. 2009, 41, 17–21. [Google Scholar] [CrossRef]
  27. Uzar, E.; Evliyaoglu, O.; Yucel, Y.; Ugur Cevik, M.; Acar, A.; Guzel, I.; Islamoglu, Y.; Colpan, L.; Tasdemir, N. Serum cytokine and pro-brain natriuretic peptide (BNP) levels in patients with migraine. Eur. Rev. Med. Pharmacol. Sci. 2011, 15, 1111–1116. [Google Scholar]
  28. Oliveira, A.B.; Bachi, A.L.L.; Ribeiro, R.T.; Mello, M.T.; Tufik, S.; Peres, M.F.P. Unbalanced plasma TNF-α and IL-12/IL-10 profile in women with migraine is associated with psychological and physiological outcomes. J. Neuroimmunol. 2017, 313, 138–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Duarte, H.; Teixeira, A.L.; Rocha, N.P.; Domingues, R.B. Increased interictal serum levels of CXCL8/IL-8 and CCL3/MIP-1α in migraine. Neurol. Sci. 2015, 36, 203–208. [Google Scholar] [CrossRef] [PubMed]
  30. Empl, M.; Sostak, P.; Riedel, M.; Schwarz, M.; Müller, N.; Förderreuther, S.; Straube, A. Decreased sTNF-RI in migraine patients? Cephalalgia 2003, 23, 55–58. [Google Scholar] [CrossRef]
  31. Michalak, S.; Kalinowska-Lyszczarz, A.; Wegrzyn, D.; Niezgoda, A.; Losy, J.; Osztynowicz, K.; Kozubski, W. Increased Serum CD14 Level Is Associated with Depletion of TNF-alpha in Monocytes in Migraine Patients during Interictal Period. Int. J. Mol. Sci. 2017, 18, 398. [Google Scholar] [CrossRef] [Green Version]
  32. Sarchielli, P.; Alberti, A.; Vaianella, L.; Pierguidi, L.; Floridi, A.; Mazzotta, G.; Floridi, A.; Gallai, V. Chemokine levels in the jugular venous blood of migraine without aura patients during attacks. Headache 2004, 44, 961–968. [Google Scholar] [CrossRef] [PubMed]
  33. Sarchielli, P.; Floridi, A.; Mancini, M.L.; Rossi, C.; Coppola, F.; Baldi, A.; Pini, L.A.; Calabresi, P. NF-kappaB activity and iNOS expression in monocytes from internal jugular blood of migraine without aura patients during attacks. Cephalalgia 2006, 26, 1071–1079. [Google Scholar] [CrossRef]
  34. Rozen, T.; Swidan, S.Z. Elevation of CSF Tumor Necrosis Factor α Levels in New Daily Persistent Headache and Treatment Refractory Chronic Migraine. Headache J. Head Face Pain 2007, 47, 1050–1055. [Google Scholar] [CrossRef] [PubMed]
  35. Bø, S.H.; Davidsen, E.M.; Gulbrandsen, P.; Dietrichs, E.; Bovim, G.; Stovner, L.J.; White, L.R. Cerebrospinal fluid cytokine levels in migraine, tension-type headache and cervicogenic headache. Cephalalgia 2009, 29, 365–372. [Google Scholar] [CrossRef] [PubMed]
  36. Gormley, P.; Anttila, V.; Winsvold, B.S.; Palta, P.; Esko, T.; Pers, T.H.; Farh, K.H.; Cuenca-Leon, E.; Muona, M.; Furlotte, N.A.; et al. Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat. Genet. 2016, 48, 856–866. [Google Scholar] [CrossRef] [Green Version]
  37. Yilmaz, I.A.; Ozge, A.; Erdal, M.E.; Edgünlü, T.G.; Cakmak, S.E.; Yalin, O.O. Cytokine polymorphism in patients with migraine: Some suggestive clues of migraine and inflammation. Pain Med. 2010, 11, 492–497. [Google Scholar] [CrossRef] [Green Version]
  38. Perry, C.J.; Blake, P.; Buettner, C.; Papavassiliou, E.; Schain, A.J.; Bhasin, M.K.; Burstein, R. Upregulation of inflammatory gene transcripts in periosteum of chronic migraineurs: Implications for extracranial origin of headache. Ann. Neurol. 2016, 79, 1000–1013. [Google Scholar] [CrossRef] [Green Version]
  39. Somjen, G.G.; Aitken, P.G.; Czéh, G.L.; Herreras, O.; Jing, J.; Young, J.N. Mechanism of spreading depression: A review of recent findings and a hypothesis. Can. J. Physiol. Pharmacol. 1992, 70, S248–S254. [Google Scholar] [CrossRef]
  40. Somjen, G.G. Mechanisms of spreading depression and hypoxic spreading depression-like depolarization. Physiol. Rev. 2001, 81, 1065–1096. [Google Scholar] [CrossRef] [Green Version]
  41. Leao, A.A. Further observations on the spreading depression of activity in the cerebral cortex. J. Neurophysiol. 1947, 10, 409–414. [Google Scholar] [CrossRef]
  42. Dreier, J.P.; Fabricius, M.; Ayata, C.; Sakowitz, O.W.; Shuttleworth, C.W.; Dohmen, C.; Graf, R.; Vajkoczy, P.; Helbok, R.; Suzuki, M.; et al. Recording, analysis, and interpretation of spreading depolarizations in neurointensive care: Review and recommendations of the COSBID research group. J. Cereb. Blood Flow Metab. 2017, 37, 1595–1625. [Google Scholar] [CrossRef] [PubMed]
  43. Hartings, J.A.; Shuttleworth, C.W.; Kirov, S.A.; Ayata, C.; Hinzman, J.M.; Foreman, B.; Andrew, R.D.; Boutelle, M.G.; Brennan, K.C.; Carlson, A.P.; et al. The continuum of spreading depolarizations in acute cortical lesion development: Examining Leão’s legacy. J. Cereb. Blood Flow Metab. 2017, 37, 1571–1594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Zhang, X.; Levy, D.; Kainz, V.; Noseda, R.; Jakubowski, M.; Burstein, R. Activation of central trigeminovascular neurons by cortical spreading depression. Ann. Neurol. 2011, 69, 855–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ayata, C.; Jin, H.; Kudo, C.; Dalkara, T.; Moskowitz, M.A. Suppression of cortical spreading depression in migraine prophylaxis. Ann. Neurol. 2006, 59, 652–661. [Google Scholar] [CrossRef] [PubMed]
  46. Karatas, H.; Erdener, S.E.; Gursoy-Ozdemir, Y.; Lule, S.; Eren-Koçak, E.; Sen, Z.D.; Dalkara, T. Spreading depression triggers headache by activating neuronal Panx1 channels. Science 2013, 339, 1092–1095. [Google Scholar] [CrossRef] [PubMed]
  47. Kraig, R.P.; Mitchell, H.M.; Christie-Pope, B.; Kunkler, P.E.; White, D.M.; Tang, Y.P.; Langan, G. TNF-alpha and Microglial Hormetic Involvement in Neurological Health & Migraine. Dose Response 2010, 8, 389–413. [Google Scholar] [CrossRef]
  48. Levy, D. Endogenous mechanisms underlying the activation and sensitization of meningeal nociceptors: The role of immuno-vascular interactions and cortical spreading depression. Curr. Pain Headache Rep. 2012, 16, 270–277. [Google Scholar] [CrossRef]
  49. Jander, S.; Schroeter, M.; Peters, O.; Witte, O.W.; Stoll, G. Cortical spreading depression induces proinflammatory cytokine gene expression in the rat brain. J. Cereb. Blood Flow Metab. 2001, 21, 218–225. [Google Scholar] [CrossRef] [Green Version]
  50. Ghaemi, A.; Sajadian, A.; Khodaie, B.; Lotfinia, A.A.; Lotfinia, M.; Aghabarari, A.; Khaleghi Ghadiri, M.; Meuth, S.; Gorji, A. Immunomodulatory Effect of Toll-Like Receptor-3 Ligand Poly I:C on Cortical Spreading Depression. Mol. Neurobiol. 2016, 53, 143–154. [Google Scholar] [CrossRef]
  51. Ghaemi, A.; Alizadeh, L.; Babaei, S.; Jafarian, M.; Khaleghi Ghadiri, M.; Meuth, S.G.; Kovac, S.; Gorji, A. Astrocyte-mediated inflammation in cortical spreading depression. Cephalalgia 2018, 38, 626–638. [Google Scholar] [CrossRef] [PubMed]
  52. Kunkler, P.E.; Hulse, R.E.; Kraig, R.P. Multiplexed cytokine protein expression profiles from spreading depression in hippocampal organotypic cultures. J. Cereb. Blood Flow Metab. 2004, 24, 829–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Thompson, C.S.; Hakim, A.M. Cortical spreading depression modifies components of the inflammatory cascade. Mol. Neurobiol. 2005, 32, 51–57. [Google Scholar] [CrossRef]
  54. Chen, S.P.; Qin, T.; Seidel, J.L.; Zheng, Y.; Eikermann, M.; Ferrari, M.D.; van den Maagdenberg, A.; Moskowitz, M.A.; Ayata, C.; Eikermann-Haerter, K. Inhibition of the P2X7-PANX1 complex suppresses spreading depolarization and neuroinflammation. Brain 2017, 140, 1643–1656. [Google Scholar] [CrossRef] [PubMed]
  55. Choudhuri, R.; Cui, L.; Yong, C.; Bowyer, S.; Klein, R.M.; Welch, K.M.; Berman, N.E. Cortical spreading depression and gene regulation: Relevance to migraine. Ann. Neurol. 2002, 51, 499–506. [Google Scholar] [CrossRef]
  56. Urbach, A.; Bruehl, C.; Witte, O.W. Microarray-based long-term detection of genes differentially expressed after cortical spreading depression. Eur. J. Neurosci. 2006, 24, 841–856. [Google Scholar] [CrossRef]
  57. Wang, H.; Peca, J.; Matsuzaki, M.; Matsuzaki, K.; Noguchi, J.; Qiu, L.; Wang, D.; Zhang, F.; Boyden, E.; Deisseroth, K.; et al. High-speed mapping of synaptic connectivity using photostimulation in Channelrhodopsin-2 transgenic mice. Proc. Natl. Acad. Sci. USA 2007, 104, 8143–8148. [Google Scholar] [CrossRef] [Green Version]
  58. Arenkiel, B.R.; Peca, J.; Davison, I.G.; Feliciano, C.; Deisseroth, K.; Augustine, G.J.; Ehlers, M.D.; Feng, G. In vivo light-induced activation of neural circuitry in transgenic mice expressing channelrhodopsin-2. Neuron 2007, 54, 205–218. [Google Scholar] [CrossRef] [Green Version]
  59. Chung, D.Y.; Sadeghian, H.; Qin, T.; Lule, S.; Lee, H.; Karakaya, F.; Goins, S.; Oka, F.; Yaseen, M.A.; Houben, T.; et al. Determinants of Optogenetic Cortical Spreading Depolarizations. Cereb. Cortex 2019, 29, 1150–1161. [Google Scholar] [CrossRef]
  60. Takizawa, T.; Qin, T.; Lopes de Morais, A.; Sugimoto, K.; Chung, J.Y.; Morsett, L.; Mulder, I.; Fischer, P.; Suzuki, T.; Anzabi, M.; et al. Non-invasively triggered spreading depolarizations induce a rapid pro-inflammatory response in cerebral cortex. J. Cereb. Blood Flow Metab. 2019, 40, 1117–1131. [Google Scholar] [CrossRef]
  61. Friedman, B.W.; Greenwald, P.; Bania, T.C.; Esses, D.; Hochberg, M.; Solorzano, C.; Corbo, J.; Chu, J.; Chew, E.; Cheung, P.; et al. Randomized trial of IV dexamethasone for acute migraine in the emergency department. Neurology 2007, 69, 2038–2044. [Google Scholar] [CrossRef]
  62. Rowe, B.H.; Colman, I.; Edmonds, M.L.; Blitz, S.; Walker, A.; Wiens, S. Randomized controlled trial of intravenous dexamethasone to prevent relapse in acute migraine headache. Headache 2008, 48, 333–340. [Google Scholar] [CrossRef] [PubMed]
  63. Donaldson, D.; Sundermann, R.; Jackson, R.; Bastani, A. Intravenous dexamethasone vs placebo as adjunctive therapy to reduce the recurrence rate of acute migraine headaches: A multicenter, double-blinded, placebo-controlled randomized clinical trial. Am. J. Emerg. Med. 2008, 26, 124–130. [Google Scholar] [CrossRef]
  64. Demartini, C.; Greco, R.; Zanaboni, A.M.; Sances, G.; De Icco, R.; Borsook, D.; Tassorelli, C. Nitroglycerin as a comparative experimental model of migraine pain: From animal to human and back. Prog. Neurobiol. 2019, 177, 15–32. [Google Scholar] [CrossRef]
  65. Reuter, U.; Bolay, H.; Jansen-Olesen, I.; Chiarugi, A.; Sanchez del Rio, M.; Letourneau, R.; Theoharides, T.C.; Waeber, C.; Moskowitz, M.A. Delayed inflammation in rat meninges: Implications for migraine pathophysiology. Brain 2001, 124, 2490–2502. [Google Scholar] [CrossRef] [PubMed]
  66. Reuter, U.; Chiarugi, A.; Bolay, H.; Moskowitz, M.A. Nuclear factor-kappaB as a molecular target for migraine therapy. Ann. Neurol. 2002, 51, 507–516. [Google Scholar] [CrossRef]
  67. Greco, R.; Demartini, C.; Zanaboni, A.M.; Redavide, E.; Pampalone, S.; Toldi, J.; Fülöp, F.; Blandini, F.; Nappi, G.; Sandrini, G.; et al. Effects of kynurenic acid analogue 1 (KYNA-A1) in nitroglycerin-induced hyperalgesia: Targets and anti-migraine mechanisms. Cephalalgia 2017, 37, 1272–1284. [Google Scholar] [CrossRef] [PubMed]
  68. Greco, R.; Demartini, C.; Zanaboni, A.; Casini, I.; De Icco, R.; Reggiani, A.; Misto, A.; Piomelli, D.; Tassorelli, C. Characterization of the peripheral FAAH inhibitor, URB937, in animal models of acute and chronic migraine. Neurobiol. Dis. 2021, 147, 105157. [Google Scholar] [CrossRef]
  69. He, W.; Long, T.; Pan, Q.; Zhang, S.; Zhang, Y.; Zhang, D.; Qin, G.; Chen, L.; Zhou, J. Microglial NLRP3 inflammasome activation mediates IL-1β release and contributes to central sensitization in a recurrent nitroglycerin-induced migraine model. J. Neuroinflammation 2019, 16, 78. [Google Scholar] [CrossRef] [Green Version]
  70. Sparaco, M.; Feleppa, M.; Lipton, R.B.; Rapoport, A.M.; Bigal, M.E. Mitochondrial dysfunction and migraine: Evidence and hypotheses. Cephalalgia 2006, 26, 361–372. [Google Scholar] [CrossRef]
  71. Yilmaz, G.; Sürer, H.; Inan, L.E.; Coskun, O.; Yücel, D. Increased nitrosative and oxidative stress in platelets of migraine patients. Tohoku J. Exp. Med. 2007, 211, 23–30. [Google Scholar] [CrossRef] [Green Version]
  72. Rajamäki, K.; Nordström, T.; Nurmi, K.; Åkerman, K.E.; Kovanen, P.T.; Öörni, K.; Eklund, K.K. Extracellular acidosis is a novel danger signal alerting innate immunity via the NLRP3 inflammasome. J. Biol. Chem. 2013, 288, 13410–13419. [Google Scholar] [CrossRef] [Green Version]
  73. Gölöncsér, F.; Sperlágh, B. Effect of genetic deletion and pharmacological antagonism of P2X7 receptors in a mouse animal model of migraine. J. Headache Pain 2014, 15, 24. [Google Scholar] [CrossRef] [Green Version]
  74. Gursoy-Ozdemir, Y.; Qiu, J.; Matsuoka, N.; Bolay, H.; Bermpohl, D.; Jin, H.; Wang, X.; Rosenberg, G.A.; Lo, E.H.; Moskowitz, M.A. Cortical spreading depression activates and upregulates MMP-9. J. Clin. Investig. 2004, 113, 1447–1455. [Google Scholar] [CrossRef]
  75. Cottier, K.E.; Galloway, E.A.; Calabrese, E.C.; Tome, M.E.; Liktor-Busa, E.; Kim, J.; Davis, T.P.; Vanderah, T.W.; Largent-Milnes, T.M. Loss of Blood-Brain Barrier Integrity in a KCl-Induced Model of Episodic Headache Enhances CNS Drug Delivery. eNeuro 2018, 5. [Google Scholar] [CrossRef] [PubMed]
  76. Sadeghian, H.; Lacoste, B.; Qin, T.; Toussay, X.; Rosa, R.; Oka, F.; Chung, D.Y.; Takizawa, T.; Gu, C.; Ayata, C. Spreading depolarizations trigger caveolin-1-dependent endothelial transcytosis. Ann. Neurol. 2018, 84, 409–423. [Google Scholar] [CrossRef] [PubMed]
  77. Harriott, A.M.; Takizawa, T.; Chung, D.Y.; Chen, S.P. Spreading depression as a preclinical model of migraine. J. Headache Pain 2019, 20, 45. [Google Scholar] [CrossRef] [PubMed]
  78. Lundblad, C.; Haanes, K.A.; Grände, G.; Edvinsson, L. Experimental inflammation following dural application of complete Freund’s adjuvant or inflammatory soup does not alter brain and trigeminal microvascular passage. J. Headache Pain 2015, 16, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Edvinsson, L.; Tfelt-Hansen, P. The blood-brain barrier in migraine treatment. Cephalalgia 2008, 28, 1245–1258. [Google Scholar] [CrossRef]
  80. Imamura, K.; Takeshima, T.; Fusayasu, E.; Nakashima, K. Increased plasma matrix metalloproteinase-9 levels in migraineurs. Headache 2008, 48, 135–139. [Google Scholar] [CrossRef]
  81. Martins-Oliveira, A.; Gonçalves, F.M.; Speciali, J.G.; Fontana, V.; Izidoro-Toledo, T.C.; Belo, V.A.; Dach, F.; Tanus-Santos, J.E. Specific matrix metalloproteinase 9 (MMP-9) haplotype affect the circulating MMP-9 levels in women with migraine. J. Neuroimmunol. 2012, 252, 89–94. [Google Scholar] [CrossRef]
  82. Dong, L.; Qiao, H.; Zhang, X.; Zhang, X.; Wang, C.; Wang, L.; Cui, L.; Zhao, J.; Xing, Y.; Li, Y.; et al. Parthenolide is neuroprotective in rat experimental stroke model: Downregulating NF-κB, phospho-p38MAPK, and caspase-1 and ameliorating BBB permeability. Mediat. Inflamm. 2013, 2013, 370804. [Google Scholar] [CrossRef] [Green Version]
  83. Sarrazin, S.; Adam, E.; Lyon, M.; Depontieu, F.; Motte, V.; Landolfi, C.; Lortat-Jacob, H.; Bechard, D.; Lassalle, P.; Delehedde, M. Endocan or endothelial cell specific molecule-1 (ESM-1): A potential novel endothelial cell marker and a new target for cancer therapy. Biochim. Biophys. Acta 2006, 1765, 25–37. [Google Scholar] [CrossRef]
  84. Hougaard, A.; Amin, F.M.; Christensen, C.E.; Younis, S.; Wolfram, F.; Cramer, S.P.; Larsson, H.B.W.; Ashina, M. Increased brainstem perfusion, but no blood-brain barrier disruption, during attacks of migraine with aura. Brain 2017, 140, 1633–1642. [Google Scholar] [CrossRef]
  85. Amin, F.M.; Hougaard, A.; Cramer, S.P.; Christensen, C.E.; Wolfram, F.; Larsson, H.B.W.; Ashina, M. Intact blood-brain barrier during spontaneous attacks of migraine without aura: A 3T DCE-MRI study. Eur. J. Neurol. 2017, 24, 1116–1124. [Google Scholar] [CrossRef]
  86. Schankin, C.J.; Maniyar, F.H.; Seo, Y.; Kori, S.; Eller, M.; Chou, D.E.; Blecha, J.; Murphy, S.T.; Hawkins, R.A.; Sprenger, T.; et al. Ictal lack of binding to brain parenchyma suggests integrity of the blood-brain barrier for 11C-dihydroergotamine during glyceryl trinitrate-induced migraine. Brain 2016, 139, 1994–2001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Jansson, D.; Rustenhoven, J.; Feng, S.; Hurley, D.; Oldfield, R.L.; Bergin, P.S.; Mee, E.W.; Faull, R.L.; Dragunow, M. A role for human brain pericytes in neuroinflammation. J. Neuroinflamm. 2014, 11, 104. [Google Scholar] [CrossRef] [Green Version]
  88. Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef] [PubMed]
  89. Khennouf, L.; Gesslein, B.; Brazhe, A.; Octeau, J.C.; Kutuzov, N.; Khakh, B.S.; Lauritzen, M. Active role of capillary pericytes during stimulation-induced activity and spreading depolarization. Brain 2018, 141, 2032–2046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Kovac, A.; Erickson, M.A.; Banks, W.A. Brain microvascular pericytes are immunoactive in culture: Cytokine, chemokine, nitric oxide, and LRP-1 expression in response to lipopolysaccharide. J. Neuroinflammation 2011, 8, 139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Alcendor, D.J.; Charest, A.M.; Zhu, W.Q.; Vigil, H.E.; Knobel, S.M. Infection and upregulation of proinflammatory cytokines in human brain vascular pericytes by human cytomegalovirus. J. Neuroinflammation 2012, 9, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Matsumoto, J.; Takata, F.; Machida, T.; Takahashi, H.; Soejima, Y.; Funakoshi, M.; Futagami, K.; Yamauchi, A.; Dohgu, S.; Kataoka, Y. Tumor necrosis factor-α-stimulated brain pericytes possess a unique cytokine and chemokine release profile and enhance microglial activation. Neurosci. Lett. 2014, 578, 133–138. [Google Scholar] [CrossRef] [PubMed]
  93. Montagne, A.; Barnes, S.R.; Sweeney, M.D.; Halliday, M.R.; Sagare, A.P.; Zhao, Z.; Toga, A.W.; Jacobs, R.E.; Liu, C.Y.; Amezcua, L.; et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015, 85, 296–302. [Google Scholar] [CrossRef] [Green Version]
  94. Matsumoto, J.; Dohgu, S.; Takata, F.; Machida, T.; Bölükbaşi Hatip, F.F.; Hatip-Al-Khatib, I.; Yamauchi, A.; Kataoka, Y. TNF-α-sensitive brain pericytes activate microglia by releasing IL-6 through cooperation between IκB-NFκB and JAK-STAT3 pathways. Brain Res. 2018, 1692, 34–44. [Google Scholar] [CrossRef] [PubMed]
  95. Klement, W.; Garbelli, R.; Zub, E.; Rossini, L.; Tassi, L.; Girard, B.; Blaquiere, M.; Bertaso, F.; Perroy, J.; de Bock, F.; et al. Seizure progression and inflammatory mediators promote pericytosis and pericyte-microglia clustering at the cerebrovasculature. Neurobiol. Dis. 2018, 113, 70–81. [Google Scholar] [CrossRef] [PubMed]
  96. Dziewulska, D.; Kierdaszuk, B. Ultrastructural changes in microvessels in familial hemiplegic migraine with CACNA1A mutation. Clin. Neuropathol. 2018, 37, 283–287. [Google Scholar] [CrossRef]
  97. Kossoff, E.H.; Andermann, F. Migraine and Epilepsy. Semin. Pediatr. Neurol. 2010, 17, 117–122. [Google Scholar] [CrossRef]
  98. Mantegazza, M.; Cestèle, S. Pathophysiological mechanisms of migraine and epilepsy: Similarities and differences. Neurosci. Lett. 2018, 667, 92–102. [Google Scholar] [CrossRef]
  99. Cheng, J.; Korte, N.; Nortley, R.; Sethi, H.; Tang, Y.; Attwell, D. Targeting pericytes for therapeutic approaches to neurological disorders. Acta Neuropathol. 2018, 136, 507–523. [Google Scholar] [CrossRef] [Green Version]
  100. Zhang, X.; Burstein, R.; Levy, D. Local action of the proinflammatory cytokines IL-1β and IL-6 on intracranial meningeal nociceptors. Cephalalgia 2012, 32, 66–72. [Google Scholar] [CrossRef]
  101. Franceschini, A.; Vilotti, S.; Ferrari, M.D.; van den Maagdenberg, A.M.; Nistri, A.; Fabbretti, E. TNFα levels and macrophages expression reflect an inflammatory potential of trigeminal ganglia in a mouse model of familial hemiplegic migraine. PLoS ONE 2013, 8, e52394. [Google Scholar] [CrossRef]
  102. Lombardo, S.D.; Mazzon, E.; Basile, M.S.; Cavalli, E.; Bramanti, P.; Nania, R.; Fagone, P.; Nicoletti, F.; Petralia, M.C. Upregulation of IL-1 Receptor Antagonist in a Mouse Model of Migraine. Brain Sci. 2019, 9, 172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. De Corato, A.; Lisi, L.; Capuano, A.; Tringali, G.; Tramutola, A.; Navarra, P.; Russo, C.D. Trigeminal satellite cells express functional calcitonin gene-related peptide receptors, whose activation enhances interleukin-1β pro-inflammatory effects. J. Neuroimmunol. 2011, 237, 39–46. [Google Scholar] [CrossRef]
  104. Vezzani, A.; French, J.; Bartfai, T.; Baram, T.Z. The role of inflammation in epilepsy. Nat. Rev. Neurol. 2011, 7, 31–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Van Vliet, E.A.; Aronica, E.; Vezzani, A.; Ravizza, T. Review: Neuroinflammatory pathways as treatment targets and biomarker candidates in epilepsy: Emerging evidence from preclinical and clinical studies. Neuropathol. Appl. Neurobiol. 2018, 44, 91–111. [Google Scholar] [CrossRef] [PubMed]
  106. Kenney-Jung, D.L.; Vezzani, A.; Kahoud, R.J.; LaFrance-Corey, R.G.; Ho, M.L.; Muskardin, T.W.; Wirrell, E.C.; Howe, C.L.; Payne, E.T. Febrile infection-related epilepsy syndrome treated with anakinra. Ann. Neurol. 2016, 80, 939–945. [Google Scholar] [CrossRef]
  107. Jyonouchi, H.; Geng, L. Intractable Epilepsy (IE) and Responses to Anakinra, a Human Recombinant IL-1 Receptor Agonist (IL-1ra): Case Reports. J. Clin. Cell. Immunol. 2016, 7, 456. [Google Scholar] [CrossRef] [Green Version]
  108. Yamanaka, G.; Ishida, Y.; Kanou, K.; Suzuki, S.; Watanabe, Y.; Takamatsu, T.; Morichi, S.; Go, S.; Oana, S.; Yamazaki, T.; et al. Towards a Treatment for Neuroinflammation in Epilepsy: Interleukin-1 Receptor Antagonist, Anakinra, as a Potential Treatment in Intractable Epilepsy. Int. J. Mol. Sci. 2021, 22, 6282. [Google Scholar] [CrossRef]
  109. Smith, C.J.; Hulme, S.; Vail, A.; Heal, C.; Parry-Jones, A.R.; Scarth, S.; Hopkins, K.; Hoadley, M.; Allan, S.M.; Rothwell, N.J.; et al. SCIL-STROKE (Subcutaneous Interleukin-1 Receptor Antagonist in Ischemic Stroke): A Randomized Controlled Phase 2 Trial. Stroke 2018, 49, 1210–1216. [Google Scholar] [CrossRef] [Green Version]
  110. Galea, J.; Ogungbenro, K.; Hulme, S.; Patel, H.; Scarth, S.; Hoadley, M.; Illingworth, K.; McMahon, C.J.; Tzerakis, N.; King, A.T.; et al. Reduction of inflammation after administration of interleukin-1 receptor antagonist following aneurysmal subarachnoid hemorrhage: Results of the Subcutaneous Interleukin-1Ra in SAH (SCIL-SAH) study. J. Neurosurg. 2018, 128, 515–523. [Google Scholar] [CrossRef] [Green Version]
  111. Sibley, C.H.; Plass, N.; Snow, J.; Wiggs, E.A.; Brewer, C.C.; King, K.A.; Zalewski, C.; Kim, H.J.; Bishop, R.; Hill, S.; et al. Sustained response and prevention of damage progression in patients with neonatal-onset multisystem inflammatory disease treated with anakinra: A cohort study to determine three- and five-year outcomes. Arthritis Rheum. 2012, 64, 2375–2386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kullenberg, T.; Löfqvist, M.; Leinonen, M.; Goldbach-Mansky, R.; Olivecrona, H. Long-term safety profile of anakinra in patients with severe cryopyrin-associated periodic syndromes. Rheumatology 2016, 55, 1499–1506. [Google Scholar] [CrossRef] [Green Version]
  113. Oby, E.; Janigro, D. The blood-brain barrier and epilepsy. Epilepsia 2006, 47, 1761–1774. [Google Scholar] [CrossRef]
  114. Yamanaka, G.; Morichi, S.; Takamatsu, T.; Watanabe, Y.; Suzuki, S.; Ishida, Y.; Oana, S.; Yamazaki, T.; Takata, F.; Kawashima, H. Links between Immune Cells from the Periphery and the Brain in the Pathogenesis of Epilepsy: A Narrative Review. Int. J. Mol. Sci. 2021, 22, 4395. [Google Scholar] [CrossRef]
  115. Landmann, E.C.; Walker, U.A. Pharmacological treatment options for cryopyrin-associated periodic syndromes. Expert Rev. Clin. Pharmacol. 2017, 10, 855–864. [Google Scholar] [CrossRef] [PubMed]
  116. Gonçalves, N.P.; Teixeira-Coelho, M.; Saraiva, M.J. Protective role of anakinra against transthyretin-mediated axonal loss and cell death in a mouse model of familial amyloidotic polyneuropathy. J. Neuropathol. Exp. Neurol. 2015, 74, 203–217. [Google Scholar] [CrossRef] [Green Version]
  117. Gonçalves, N.P.; Vieira, P.; Saraiva, M.J. Interleukin-1 signaling pathway as a therapeutic target in transthyretin amyloidosis. Amyloid 2014, 21, 175–184. [Google Scholar] [CrossRef] [Green Version]
  118. Coll, R.C.; Robertson, A.A.; Chae, J.J.; Higgins, S.C.; Muñoz-Planillo, R.; Inserra, M.C.; Vetter, I.; Dungan, L.S.; Monks, B.G.; Stutz, A.; et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat. Med. 2015, 21, 248–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Cho, J.W.; Lee, K.S.; Kim, C.W. Curcumin attenuates the expression of IL-1beta, IL-6, and TNF-alpha as well as cyclin E in TNF-alpha-treated HaCaT cells; NF-kappaB and MAPKs as potential upstream targets. Int. J. Mol. Med. 2007, 19, 469–474. [Google Scholar]
  120. Ferreira, N.; Gonçalves, N.P.; Saraiva, M.J.; Almeida, M.R. Curcumin: A multi-target disease-modifying agent for late-stage transthyretin amyloidosis. Sci. Rep. 2016, 6, 26623. [Google Scholar] [CrossRef]
  121. Fan, C.; Song, Q.; Wang, P.; Li, Y.; Yang, M.; Yu, S.Y. Neuroprotective Effects of Curcumin on IL-1β-Induced Neuronal Apoptosis and Depression-Like Behaviors Caused by Chronic Stress in Rats. Front. Cell. Neurosci. 2018, 12, 516. [Google Scholar] [CrossRef] [PubMed]
  122. Ferreira, N.; Saraiva, M.J.; Almeida, M.R. Uncovering the Neuroprotective Mechanisms of Curcumin on Transthyretin Amyloidosis. Int. J. Mol. Sci. 2019, 20, 1287. [Google Scholar] [CrossRef] [Green Version]
  123. Bulboacă, A.E.; Bolboacă, S.D.; Stănescu, I.C.; Sfrângeu, C.A.; Bulboacă, A.C. Preemptive Analgesic and Antioxidative Effect of Curcumin for Experimental Migraine. Biomed. Res. Int. 2017, 2017, 4754701. [Google Scholar] [CrossRef] [PubMed]
  124. Chen, L.; Li, X.; Huang, L.; Wu, Q.; Chen, L.; Wan, Q. Chemical stimulation of the intracranial dura activates NALP3 inflammasome in trigeminal ganglia neurons. Brain Res. 2014, 1566, 1–11. [Google Scholar] [CrossRef] [PubMed]
Figure 1. CGRP and the trigemino-vascular system in migraine.
Figure 1. CGRP and the trigemino-vascular system in migraine.
Ijms 22 08929 g001
Figure 2. Potential for therapies using the IL-1β/IL-1R1 axis.
Figure 2. Potential for therapies using the IL-1β/IL-1R1 axis.
Ijms 22 08929 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yamanaka, G.; Suzuki, S.; Morishita, N.; Takeshita, M.; Kanou, K.; Takamatsu, T.; Suzuki, S.; Morichi, S.; Watanabe, Y.; Ishida, Y.; et al. Role of Neuroinflammation and Blood-Brain Barrier Permutability on Migraine. Int. J. Mol. Sci. 2021, 22, 8929. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168929

AMA Style

Yamanaka G, Suzuki S, Morishita N, Takeshita M, Kanou K, Takamatsu T, Suzuki S, Morichi S, Watanabe Y, Ishida Y, et al. Role of Neuroinflammation and Blood-Brain Barrier Permutability on Migraine. International Journal of Molecular Sciences. 2021; 22(16):8929. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168929

Chicago/Turabian Style

Yamanaka, Gaku, Shinji Suzuki, Natsumi Morishita, Mika Takeshita, Kanako Kanou, Tomoko Takamatsu, Shunsuke Suzuki, Shinichiro Morichi, Yusuke Watanabe, Yu Ishida, and et al. 2021. "Role of Neuroinflammation and Blood-Brain Barrier Permutability on Migraine" International Journal of Molecular Sciences 22, no. 16: 8929. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168929

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop