- Review
- Open access
- Published:
Unmasking the relationship between CGRP and glutamate: from peripheral excitation to central sensitization in migraine
The Journal of Headache and Pain volume 26, Article number: 101 (2025)
Abstract
The exact mechanisms that trigger the activation of the trigeminovascular system in migraine remain unclear. The involvement of calcitonin gene-related peptide (CGRP) in migraine is well-documented, and treatments aimed at blocking CGRP activity have proven successful in reducing migraine attacks for some patients. However, around one third of individuals do not respond to these therapies, which are also limited by factors like cost, side effects, and contraindications. There is growing evidence suggesting that glutamate, an excitatory neurotransmitter, plays a crucial role in the onset and maintenance of migraine pain, partially by enhancing CGRP release. Increased glutamate levels have been linked to both peripheral and central sensitization, potentially contributing to the development and persistence of chronic migraine. The relationship between CGRP and glutamate is complex, with glutamate possibly acting as an upstream trigger for CGRP release. This review examines the interplay between CGRP and glutamate, and their involvement in both peripheral and central sensitization. It also explores the therapeutic potential of targeting either glutamate or CGRP, aiming to address both peripheral and central migraine mechanisms. Finally, the role of triggers in migraine initiation at the peripheral level is discussed, offering insights into potential preventive strategies.
Introduction
Activation of the trigeminovascular system is a key pathological event in the initiation of a migraine attack [1]. Current evidence suggests that migraine pain originates peripherally, with the activation of nociceptive neurons innervating the dura mater [2]. However, the exact mechanisms that trigger this activation remain uncertain. Preclinical and clinical studies have identified calcitonin gene-related peptide (CGRP) as a crucial player in nociception [3, 4]. Its critical role in migraine pathophysiology is further supported by the promising results of pharmacological interventions targeting CGRP activity, which prevent migraine attacks by either blocking receptor-binding sites or modulating neuropeptide levels [5,6,7]. However, about 25–30% of patients still do not respond to these medications, leading to treatment discontinuation [8,9,10,11]. Additionally, their use is further limited by factors such as cost, side effects, and contraindications [12,13,14,15,16]. Notably, these treatments exert their effects mainly peripherally, without crossing the blood-brain barrier (BBB), which supports the idea of the peripheral origin of migraine attacks [17, 18]. While the peripheral origin of migraine is well-supported, central mechanisms should not be overlooked. Migraine attacks often begin with prodromal symptoms hours to days before headache onset, highlighting central involvement in early phases [19]. Additionally, some CGRP-targeted antibodies can cross BBB to some extent and exert central effects [20, 21].
The precise mechanisms through which CGRP contributes to the pain process remain to be fully clarified. An intriguing animal study demonstrated that while topical and systemic administration of CGRP significantly increased dural blood flow, neither method resulted in the activation or sensitization of meningeal nociceptors [22]. This suggests that CGRP’s vasodilatory effect in the meninges may be insufficient alone to trigger or sensitize nociceptors, indicating that we may be overlooking another critical element in the pain cascade. Glutamate, the brain’s primary excitatory neurotransmitter, could be this missing link [23, 24]. Animal models show that elevated glutamate levels reduce the mechanical force needed to activate trigeminovascular neurons and to increase dural blood flow [25, 26]. Known for its role in excitation, glutamate is abundant in pain pathways [23, 24] and plays a key role in initiating cortical spreading depression [26]. With the documented elevation in glutamate at both peripheral and central levels among migraine patients [27,28,29], it could be likely that glutamate could serve as a central driver in this cascade.
Glutamate and CGRP may both contribute to peripheral and central sensitization, driving not only the onset and persistence of migraine headaches, but also their progression to chronic migraine [30,31,32]. While their relationship is complex, in vitro and in vivo evidence supports the upstream effect of glutamate on CGRP release [33,34,35,36]. This raises the possibility that the current approach of targeting CGRP may interrupt the chain of events midstream, leaving the glutamate-driven upstream mechanisms unaddressed.
These insights bring up two important questions. First, by targeting CGRP peripherally, could central mechanisms persist via glutamate, potentially explaining why some patients do not respond to treatment, or why migraines are not fully eliminated even in responders? Second, could we achieve therapeutic benefits if we target glutamate’s initial impact at the peripheral level? In this review, we aim to answer these questions by exploring the relationship between CGRP and glutamate, investigating which may act as the primary driver, and examining their roles in both the peripheral and central mechanisms of migraine. Additionally, we will discuss the clinical implications and challenges of targeting these neuro-messengers and provide insights into possible treatment strategies.
The trigeminovascular system
Peripheral endings of trigeminal fibers surround the dural blood vessels, forming a network which is highly responsive to vascular and neuronal changes, often referred to as the ‘trigeminovascular system’ [37]. In animal studies, stimulation of this system has been widely utilized to investigate the headache phase of migraine [37,38,39]. While it has already been established that activation of this pathway leads to pain perception, the initial triggers for this activation remain unclear.
The meninges, muscles, and blood vessels in the head receive extensive innervation from primary afferent neurons, which include myelinated Aδ fibers and unmyelinated C fibers [40]. The cell bodies of these afferent neurons are located in the sensory ganglia, including the trigeminal ganglion (TG) and cervical dorsal root ganglion [41,42,43]. Dorsal root ganglion and TG neurons are pseudo-unipolar, with axons that split to send signals both to the central area, including the dorsal horn or trigeminal nucleus caudalis (TNC), as well as to peripheral tissues near the dura [43, 44]. The TNC is a part of the brainstem and serves as the principal region for processing facial nociceptive signals from the trigeminal nerve [45]. The trigeminal nerve comprises three branches to the forehead and face: the ophthalmic, maxillary, and mandibular nerves [46]. Initial anatomical studies showed that many of the dural vascular structures are innervated by the Aδ- and C-fibers arising from the ophthalmic branch of the trigeminal nerve [47, 48]. In contrast, the cervical dorsal root ganglia of the C1-C3 nerves primarily innervate the back of the head and upper neck [49].
Primary afferents from the dura and posterior regions of the head and neck, project to the trigeminal cervical complex, a region that encompasses the spinal trigeminal nucleus in the brainstem and spinal cord segments C1 to C3 [43, 50, 51]. Therefore, the trigeminal cervical complex functions as the primary relay center for nociceptive information within the head [52]. Research on animal models has demonstrated that stimulating dural blood vessels enhances neuronal excitability, as well as increases metabolic activity and blood flow within the trigeminal cervical complex [53,54,55]. The nociceptive pathway continues as second-order neurons in the trigeminal cervical complex project along the ascending trigeminothalamic tract to third-order neurons in the thalamus, particularly the ventroposteromedial and posterior thalamic nuclei [56]. These thalamic neurons relay pain-related sensory and discriminatory information to the primary somatosensory cortex, where the perception of pain is processed and integrated [57].
Key evidence for the trigeminal system’s role in migraine headaches is that effective treatments, like the hydrophilic drug sumatriptan, and preventive therapies, such as botulinum toxin and CGRP monoclonal antibodies, do not cross the BBB [58,59,60]. Thus, these treatments work by inhibiting nociceptive signaling from peripheral trigeminal fibers in the dura [61]. Similar to blood vessels in the dura, the TG is located outside of the BBB; therefore, all protection provided by the BBB is not present for the cell bodies of these trigeminal afferents [62] (Fig. 1). In addition to dura and TG, certain parts of the brainstem, such as the area postrema, are not protected by the BBB. In a non-human primate study by Eftekhari et al., CGRP receptor expression and binding of a CGRP receptor antagonist were observed in some of these brainstem regions [63].
Peripheral model of migraine initiation. Activated primary afferents innervating blood vessels of the dura transmit signals to the TCC, a relay center in the brainstem. The TCC also receives input from the cervical dorsal root ganglia of the C1-C3 nerves, which primarily innervate the back of the head and upper neck. Second-order neurons then relay signals from the brainstem to the thalamus, which projects them to various regions of the somatosensory cortex, where pain perception occurs. Effective medications with inability to cross BBB are believed to prevent the attack by inhibition of sensory nerves at the level of the meninges. BBB, blood-brain barrier; TCC, trigeminal cervical complex. C1-C3: cervical nerves. Created in https://BioRender.com
Glutamate: an initiator of the trigeminovascular system
Glutamate is the main excitatory neurotransmitter in the central nervous system (CNS), essential for most brain functions [64, 65], and implicated in various neurological disorders including migraine [66,67,68,69,70,71]. It is stored in synaptic vesicles via a proton gradient-dependent system, with three vesicular glutamate transporters identified so far [72]. Upon neuronal stimulation, glutamate is released into the synapse through exocytosis, briefly raising its concentration and activating glutamate receptors [72]. Although glutamate is crucial for normal signal transduction in the nervous system, excessive accumulation of glutamate in the synaptic space can lead to excitotoxicity. Excitotoxicity refers to excessive activation of glutamate receptors and sustained neuronal excitation, leading to neuronal damage and ultimately cell death [73]. Glutamate receptors are classified into two types: ionotropic and metabotropic [74]. Ionotropic glutamate receptors (iGluRs) are fast-acting, ligand-gated channels, further classified into N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA), and kainic acid receptors. These have rapid excitatory effects. Metabotropic glutamate receptors (mGluRs), on the other hand, are G-protein-coupled and include eight subtypes (mGluR1-8) grouped into three categories (I-III) based on their effects on second messengers and pharmacology. Group I is linked to slow excitation, while groups II and III are involved in slow inhibition [74].
It is widely recognized that glutamate facilitates fast synaptic transmission from primary afferents to second-order nociceptive neurons [75]. Animal and human localization studies have demonstrated that NMDA, AMPA, kainate, and mGlu receptors are present within the trigeminal system [24, 76, 77]. Also, at the level of the spinal cord, all ionotropic glutamate receptors are found in both the dorsal horn [78] and the dorsal root ganglion [79, 80]. Glutamate receptors are present not only in the trigeminal cervical complex and TG, but also in other pain-modulating brain regions such as the thalamus, hypothalamus, and periaqueductal gray [81, 82]. Specifically, high concentrations of NMDA-positive neurons are found in major structures involved in migraine pain, such as the TG, TNC, and thalamus; suggesting a possible link between NMDA receptor activity and the underlying mechanisms of migraine [81, 82]. In the trigeminal nociceptive system, glutamate stored in small synaptic vesicles can be released by presynaptic spikes, activating AMPA and NMDA receptors on the postsynaptic nociceptive neurons in the TNC [24] or in dorsal horn neurons of the upper cervical segments [83, 84]. Applying a selective small fiber excitant (mustard oil), into the temporomandibular joint region in rats activates small trigeminal C-fibers and triggers the acute release of glutamate and aspartate in the TNC at the synapse with second-order neurons [85]. In support of glutamate’s role in migraine, the genes identified with familial hemiplegic migraine all encode proteins that regulate glutamate availability at synaptic terminals, ultimately enhancing neuronal excitability [86].
Peripheral trigeminal nociceptive signaling from the meninges is now regarded as a primary mechanism underlying migraine pain [2]. Interestingly, in addition to glutamate’s well-known role in transmitting nociceptive signals from primary afferents to the central area of the brain, previous findings support the role of glutamate as a peripheral pro-nociceptive agent in the trigeminal system [87,88,89]. The presence of NMDA receptors has been observed in nerve fibers that innervate dural blood vessels [90]. In line with the anatomical findings, the role of NMDA receptors in the activation of trigeminal afferents that innervate the meninges, has also been reported [90, 91]. In an interesting study by Guerrero et al., they revealed that glutamate and aspartate (both activators of NMDA receptors) successfully activated isolated TG neurons, suggesting functional NMDA receptor expression in first-order nociceptors [91]. Additional evidence supporting the role of peripheral glutamatergic system dysregulation in migraine development comes from studies showing elevated plasma glutamate levels in individuals with migraine [27, 28, 92]. This increase is present both during migraine attacks and in interictal periods [28, 93], impacting individuals with and without aura [93, 94].
Glutamate and vasodilation
Interestingly, glutamate can cause vasodilation, a phenomenon commonly related to migraine, which for many years was believed to be the main cause of migraine. The connection between glutamate and nitric oxide (NO) was first suggested when it was observed that treatment with glutamate or NMDA led to the release of NO and cyclic guanosine monophosphate (cGMP) in cerebellar cultures [95]. Furthermore, activation of NMDA receptors increases cGMP levels in the brain, a process that can be blocked by nitric oxide synthase inhibitors and NO scavengers, indicating that NO plays a downstream signaling role after NMDA receptor activation [95]. In neurons, Ca2+ influx resulting from NMDA receptor activation stimulates nitric oxide synthase, which is physically coupled to NMDA receptors [96]. Additionally, glutamate can activate NMDA receptors on endothelial cells in capillaries, triggering nitric oxide synthase induction and NO release, which promotes vasodilation [97].
CGRP: a vasodilator and amplifier in the trigeminovascular system
Calcitonin gene-related peptide (CGRP) is a 37-amino acid neuropeptide that functions as a neuromodulator. Both preclinical and clinical research indicates that CGRP is closely linked to brain nociception, though the precise mechanisms remain uncertain [98, 99]. However, it is well-established that CGRP can act as a powerful vasodilator [100]. CGRP receptors are G-protein coupled receptors and when CGRP binds to these receptors on artery smooth muscle cells, it triggers an increase in intracellular cyclic adenosine monophosphate (cAMP), leading to muscle relaxation and vasodilation [101].
The CGRP receptor is found on dural vascular smooth muscle cells, neurons, and satellite glial cells in the TG, TNC, and dorsal horns of the spinal cord [102, 103]. More specifically, they are present on Aδ fibers which are large-diameter myelinated TG neurons that do not produce CGRP, as well as on surrounding satellite glial cells [104]. In this regard, meningeal Aδ fibers possess CGRP receptors, whereas meningeal C fibers do not [105]. Unlike the CGRP receptor, CGRP itself is almost exclusively expressed in neurons, especially in sensory nerves [104]. CGRP is predominantly synthesized in unmyelinated C fibers within the trigeminovascular system [105]. These CGRP-expressing fibers are afferent (meaning that signaling is coming from the periphery and moving toward the brain), originating from the dorsal root ganglion and the TG, reaching both intracranial and extracranial regions of the head, including the dura [106]. These sensory fibers are polymodal in nature as they can be activated with thermal, chemical, and high-threshold mechanical stimuli [107]. Therefore, in response to the mentioned stimuli, a local axon reflex occurs, allowing these sensory fibers to release CGRP [108]. As mentioned earlier, dorsal root ganglion and TG neurons are pseudo-unipolar, with axons that split to send signals both to the central area, including the dorsal horn or TNC, and to peripheral tissues [44]. This distinctive structure enables the release of neurotransmitters/neuromodulators at both central and peripheral sites, facilitating two-way communication between the terminals [44].
The direct role of CGRP in activating meningeal afferents has been disproven to some extent. When CGRP is applied locally to the rodent dura mater, it does not induce firing or immediate sensitization of meningeal afferents, though cAMP analogs can effectively sensitize them [22, 109]. This lack of activation likely results from the absence of functional CGRP receptors on nerve fibers innervating the dura mater. As Lennerz et al. showed in the cranial dura mater, immunoreactivity for CGRP receptor components was observed in arterial blood vessels, mononuclear cells, and Schwann cells, but was absent in sensory axons [110]. It is noteworthy that an intriguing animal study demonstrated that CGRP preferentially activates and sensitizes trigeminal nociceptors in the dura of female mice, but not male mice [111]. Similarly, in another rodent model, intraspinal CGRP administration caused prolonged, dose-dependent mechanical hypersensitivity in female mice, whereas its effects in males were more transient [112]. Also, in hyperalgesic priming induced by interleukin-6, the CGRP receptor antagonist olcegepant, when administered intraspinally, blocked and reversed hyperalgesic priming exclusively in females [112]. These sex-specific effects of CGRP at both peripheral and central levels warrant further investigation.
Even direct injections of CGRP or its receptor antagonists into the rat TG, fail to alter the activity or mechanical sensitivity in neurons within the spinal trigeminal nucleus that receives meningeal input [113]. However, systemic administration and iontophoretic application of CGRP receptor antagonists in the spinal trigeminal nucleus effectively reduces spinal trigeminal activity [98], suggesting that CGRP primarily activates trigeminovascular neurons at their synapses within the trigeminal nucleus [114]. Direct injection of CGRP receptor antagonists into the periaqueductal gray inhibited nociceptive trigeminovascular activation, suggesting that central descending mechanisms from this region play a role in the action of CGRP and its receptor antagonists in controlling head pain [115]. This lack of a direct hyperalgesic effect may be considered paradoxical given the effectiveness of peripheral antibodies targeting CGRP and/or its receptor. It is possible that CGRP indirectly modulates central trigeminal nociceptive transmission, likely through other mediators. It should be noted that genetic mapping studies have not linked migraine to variations in the CGRP gene [116], and a genome-wide association study has similarly failed to implicate the CGRP or CGRP receptor genes [117]. Additionally, the infusion of other chemicals, such as glyceryl trinitrate, histamine, and pituitary adenylate cyclase-activating peptide, can induce delayed migraine-like headaches in migraineurs, similar to CGRP [118,119,120,121]. Overall, it is reasonable to conclude that CGRP plays a significant role in migraine pathophysiology, but it is certainly not the sole contributor.
CGRP and vasodilation
Release of CGRP from bidirectional trigeminal afferent C-fibers can activate CGRP receptors on nearby afferent Aδ fibers, which extend back to the TNC [44]. In the peripheral nervous system, CGRP’s strong vasodilatory effect can expand meningeal arteries [100]. However, the idea that dural vasodilation directly causes pain is questioned for a few reasons. First, not all substances that dilate blood vessels induce migraine pain in humans; for instance, vasoactive intestinal peptide can cause cranial vasodilation without triggering migraine in migraineurs [122]. However, it should be mentioned that there are reports of headache induction by vasoactive intestinal peptide in healthy individuals and migraine patients without aura [123, 124]. Second, some migraine-inducing substances, despite their well-known vasodilatory properties, can trigger migraines independently of significant vasodilation. For example, a 3T magnetic resonance angiography study on migraine patients investigated vascular changes during nitroglycerin-induced migraine. The results showed no significant differences in the diameters of intracerebral arteries or extracerebral arteries compared to baseline. Additionally, blood vessel diameters remained consistent between headache and non-headache sides. Blood flow in the basilar and internal carotid arteries also remained unchanged during nitroglycerin infusion or later during migraine [125]. In a cross-sectional study of migraine patients examining within subject differences in the circumference of extracranial and intracranial arterial segments between attack and attack-free days, as well as between the pain and non-pain sides, migraine pain was not associated with extracranial arterial dilation and showed only slight intracranial dilation [126]. While both topical and systemic CGRP administration led to dilation of meningeal blood vessels, it did not trigger or sensitize meningeal pain receptors in the animal model [22]. In a double-blind, crossover study, 12 patients with migraine without aura received an intravenous infusion of either human α-CGRP or a placebo. The author concluded that while CGRP induced cerebral artery dilation, the extent of this effect was minimal, making it unlikely to be the sole mechanism underlying CGRP-induced migraine [127]. There are multiple potential scenarios which may be at play here. (1) Vasodilation alone may not activate the migraine pain pathway and may simply occur alongside meningeal nociceptor activation and the release of vasoactive peptides into meningeal blood vessels. (2) A greater amount of vasodilation may be necessary to induce pain, requiring a specific threshold to be reached. (3) Neuropeptide-induced vasodilation may trigger pain transmission when peripheral sensitization has already occurred in the primary afferent. (4) Different mechanosensitivity of nociceptors may alter response to vasodilation affecting the pain pathway, for example, Aδ fibers have greater mechanosensitivity than that of C-fibers [128]. In this case, vasodilation, which causes mechanical stretching of the blood vessel walls, is likely to have a stronger effect on activating Aδ fibers compared to C-fibers. In this latter case, blocking CGRP vasodilatory action could have a more pronounced effect on Aδ fibers, as CGRP receptors are present on vascular smooth muscle cells [105].
Although evidence has questioned whether dural vasodilation directly triggers pain, it may still influence trigeminal primary afferent nociceptors. One proposed mechanism for how vascular signaling contributes to pain perception involves activation and sensitization through increased extracellular cations, primarily potassium [129]. It has been suggested that activation of either cAMP-mediated pathways (e.g., through CGRP) or cGMP-mediated pathways (e.g., through sildenafil) opens ATP-sensitive potassium channels on vascular smooth muscle cells in the walls of intracranial arteries, leading to vasodilation. This process results in the accumulation of positively charged ions in the extracellular space, creating an electrical gradient that drives these ions into neighboring trigeminal pain fibers, activating them [41]. Taken together, vasodilation is an important phenomenon observed during migraine, and its role in migraine pathophysiology should not be overlooked. However, some evidence continues to question whether it is the primary driver of pain. Instead, migraine pain may arise from a combination of vascular and neuronal factors, warranting further investigation into how these processes interact.
Studies exploring the relationship between glutamate and CGRP
The association between CGRP and glutamate has been mostly investigated in animal models so far (Table 1). The relationship between these two compounds demonstrates both upstream and downstream effects of glutamate that may be causing or mediating the effects of CGRP on migraine.
Glutamate’s upstream role in CGRP release
An in vivo model in rats showed that increased glutamate levels in peripheral tissues like the temporalis muscle led to CGRP release [35]. This is in line with findings by Benbow et al. showing that administration of 1000 mg/kg monosodium glutamate (MSG) to rats, produced a prolonged increase in plasma glutamate and CGRP concentrations, followed by headache and nausea-like behaviors [33]. These findings make sense when considering the defensive role of the BBB, where individuals with an intact BBB have lower transport of glutamate from the blood into the brain [130]. As shown by Price et al., significantly elevated serum levels of glutamate or aspartate led to a notable net increase in the concentration of these amino acids in circumventricular organ regions of the brain, like the hypothalamus, which are thought to have little to no BBB protection; while no significant increase was observed in non-circumventricular organ regions [131]. Thus, in the case of a person having normal BBB function, glutamate can only exert its initiating nociception at the dura and TG levels. However, situations which cause BBB permeability, such as head injury [132], infection [133], stress [134], high substance P (SP) levels [135], neurotoxic exposure [136], and presence of excitotoxicity itself [137], could then allow glutamate to act both peripherally and centrally.
In the brainstem, glutamate receptor antagonists inhibit CGRP release, particularly through AMPA receptors, suggesting that glutamate receptor activation is required to stimulate CGRP release centrally [36]. In the study by Kageneck [34], they hypothesized that capsaicin-induced glutamate release may facilitate CGRP release that depends on Ca2+ through presynaptic glutamate auto-receptors. Ca2+-permeable NMDA receptors could further increase Ca2+ levels in central afferent terminals, adding to the Ca2+ already introduced upon capsaicin stimulation via other channels. Interestingly, glutamate receptor antagonists significantly inhibited CGRP release in the brainstem, but only slightly in the TG, and not at all in the dura mater. This may imply a central mechanism where brainstem activation (via glutamate signaling) enhances CGRP release, influencing central pain processing in migraine.
CGRP’s downstream effect on glutamate release and neuronal excitability
In the study by Kangrga et al., CGRP perfusion increased the basal release of glutamate and aspartate from rat spinal cord slices [138]. However, this CGRP-induced release was not affected by capsaicin treatment. Since capsaicin administration selectively destroys small neurons in the dorsal root ganglia, many of which contain neuropeptides such as SP and CGRP, this finding suggests that CGRP does not stimulate glutamate and aspartate release from capsaicin-sensitive primary afferents. Instead, the source of these excitatory neurotransmitters may be local interneurons or neighboring microglial cells. Similarly, in the study by Takhshid et al., CGRP was found to enhance K+-stimulated release of aspartate and glutamate from the dorsal spinal cord [139].
Recent research has shown that CGRP in the spinal cord is not only derived from primary afferents but also from a distinct group of excitatory interneurons in the spinal cord dorsal horn and TNC [140]. Under resting conditions, CGRP interneurons are regulated by tonic inhibitory control. However, stimulating the dorsal roots through electrical activation of non-nociceptive Aβ fibers (large myelinated afferents) led to depolarization of these neurons. Additionally, chemogenetic activation of CGRP interneurons resulted in mechanical hypersensitivity. Based on this finding the authors suggested that, under certain conditions, CGRP interneurons become hyperexcitable and may contribute to either ascending pathways from the deep dorsal horn or to reflex circuits [140]. This matter needs more investigation in regard to migraine pathophysiology.
Additionally, CGRP sensitizes neurons to glutamate by increasing the density of glutamate receptors, including NMDA and AMPA receptors. Studies indicate that applying CGRP to dorsal horn neurons can alter their membrane potential [141], leading to an increase in spontaneous discharges as well as discharges triggered by intracellular depolarizing pulses [141, 142]. This heightened excitability in the neuronal membrane could, therefore, increase firing in response to NMDA and AMPA application. However, it is worth noting that in the Ebersberger study, they did not observe neuron firing induced solely by CGRP, which implies that any depolarization effect from CGRP might have been minimal, if present at all [143]. Another possible scenario is that CGRP increases Ca2+ uptake in depolarized synaptosomes [144], which may, in turn, trigger the release of glutamate [145, 146]. This CGRP-driven increase in glutamate within the tissue could cause neuronal depolarization, potentially amplifying their responsiveness to externally applied NMDA and AMPA.
Potential feedback loop
Based on the evidence reviewed, it appears that elevated glutamate levels lead to the release of CGRP, especially in the brainstem. This release, in turn, increases both glutamate release and the sensitivity of its receptors. This creates a cyclical interaction where glutamate and CGRP enhance each other’s effects, resulting in a sustained state of neuronal excitation and sensitization (Fig. 2).
Interaction Between Glutamate and CGRP Influenced by Peripheral Glutamate. (1) High levels of glutamate in the dura or blood vessels of the TG can more easily enter the brain due to the lack of BBB protection in these areas. (2) This glutamate binds to NMDA, AMPA, or kainate receptors at the peripheral side (only NMDA is shown in the figure), increasing Ca2+ influx and activating primary afferent neurons. (3) The signal is transmitted to second-order neurons in the TCC of the brainstem through glutamate released from the central end into the synaptic cleft. (4) The excitatory signal then propagates to higher brain regions involved in pain processing, such as the thalamus. (5) In addition to glutamate, CGRP is released from the central terminals of primary C fibers and activates its receptors on second-order neurons. (6) Activated second-order neurons release CGRP, which stimulates its receptors on nearby microglial cells. (7) Microglial cells, in response to CGRP receptor activation, release additional glutamate and inflammatory cytokines. This glutamate ensures the activation of second-order neurons and amplifies sensitization. TG: trigeminal ganglion, BBB: blood brain barrier, Ca2+: calcium, TCC: trigeminal cervical complex. CGRP: calcitonin gene-related peptide, NMDA: N-methyl-D-aspartic acid, AMPA: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, Created in https://BioRender.com
Role of glutamate and CGRP in development of peripheral sensitization
Peripheral sensitization related to migraine refers to a lowered activation threshold and an amplified response of sensory nerves in meninges and TG to external stimuli [147]. This is characterized by enhanced stimulus-dependent pain, known as primary hyperalgesia which is clinically evident where the headache worsens with coughing or physical activity, and through the characteristic throbbing or pulsating nature of migraine headache [148].
Glutamate’s role in peripheral sensitization
Peripheral sensitization typically occurs following peripheral nerve injury or inflammation [149]. Both of these conditions have been shown to cause an increased release of glutamate in the TG neurons [150, 151]. Glutamate can enhance inflammation at the peripheral level by directly affecting satellite glial cells [152]. These cells, which surround the cell bodies of neurons in the TG, release substances that can affect adjacent neurons, potentially altering their excitability [153]. These satellite glial cells have been shown to also release glutamate themselves, including during nociceptive signaling [152]. Depolarization of these glial cells triggers the vesicular release of glutamate, which can diffuse and influence TG neurons [152]. Activation of type C fibers in the TG leads to the release of CGRP [40]. The satellite glial cells contain CGRP receptors and can respond to CGRP by further releasing glutamate and inflammatory cytokines [154]. This process ensures the sustained release of glutamate and CGRP, contributing to peripheral sensitization. It is also important to note that exogenous administration of monosodium glutamate (MSG) can similarly activate the TG [154], which may help explain the common report from migraineurs of sensitivity to MSG as a dietary trigger for migraine [155,156,157].
CGRP’s role in peripheral sensitization
As mentioned earlier, CGRP alone may not directly trigger nociception, but evidence supports CGRP’s role in peripheral sensitization. For instance, repeated injections of low doses of CGRP into rat paws induced peripheral sensitization, as evidenced by a significant decrease in the response threshold to noxious mechanical stimuli [158]. In another study, injection of a CGRP receptor antagonist prevented the behavioral signs of hyperalgesia in rats with peripheral nerve injury [32]. Nerve damage or inflammation in the periphery leads to the localized release of proinflammatory mediators such as bradykinin, histamine, chemokines, cytokines, and glutamate [159]. These substances can sensitize peripheral nociceptors by lowering their activation threshold or directly triggering action potentials, thereby activating them. Persistent activation of the nociceptors over time, causes translational changes including- but not limited to- an increase in the production of SP and CGRP, thus maintaining a state of peripheral sensitization [159].
In animal models of migraine, it is well-established that SP and CGRP induce neurogenic inflammation, characterized by vasodilation, plasma protein extravasation (from leaky blood vessels), and mast cell degranulation. This mechanism is a key piece of evidence supporting the involvement of CGRP in migraine pathophysiology. However, some findings in humans cannot be overlooked. In an attempt to explore the acute effects of neuropeptides on human skin, SP induced both vasodilation and plasma protein extravasation, while CGRP caused potent vasodilation without triggering plasma extravasation [160]. In another study, intense electrical stimulation of cutaneous nerves in human volunteers, which triggered the release of SP and CGRP, failed to induce mast cell degranulation, histamine release, or plasma protein extravasation [161]. It should be noted that research on SP has primarily focused on a single receptor pathway (neurokinin-1) [162]. However, it is possible that SP may also act through other receptors, contributing to neurogenic inflammation and, consequently, migraine. Overall, the role of neurogenic inflammation in migraine pathophysiology is well-supported, particularly in animal models (as detailed in the review by Spekker et al. [163]); However, further clinical studies are needed to fully validate this theory in clinical models.
On the other hand, the release of inflammatory cytokines as a result of these mediators can lead to BBB permeability [164]. There is also evidence that SP, as mentioned earlier, can also cause a temporary disruption in BBB integrity in in vitro studies [135]. Compromising the BBB could be an example of how peripheral sensitization can start transitioning into central sensitization.
Role of glutamate and CGRP in development of central sensitization
Central sensitization happens after peripheral sensitization and is thought to be the underlying mechanism of chronic migraine; clinically presenting as allodynia [165]. Allodynia occurs when a typically non-painful stimulus triggers pain, with sensitivity extending beyond the original pain location [166, 167]. Around 80% of migraine sufferers develop cutaneous allodynia [168, 169]. Central sensitization differs from peripheral sensitization by involving hyperexcitability of second-order neurons in the trigeminocervical complex or higher-order neurons in the thalamus, even in non-inflamed tissues [170].
Glutamate’s role in central sensitization
Central sensitization is believed to be mainly driven by the increased activation of NMDA glutamate receptors on higher-order neurons [171]. Studies have indicated that administering glutamate or its receptor agonists induces allodynia and hyperalgesia [172, 173]. Conversely, blocking glutamate counteracts these nociceptive effects [174, 175]. In normal situations, magnesium (Mg2+) serves as a block of the NMDA receptor. When nociceptor-evoked synaptic potentials accumulate, they cause membrane depolarization, which removes the voltage-dependent Mg2+ block from NMDA channels, allowing Ca2+ to enter the cell [176]. This influx of Ca2+ activates intracellular kinases, triggering post-translational modifications, particularly the phosphorylation of membrane receptors and ion channels. As a result, there is an increase in neuronal excitability, which makes the neurons more sensitive to inputs that would usually fall below the threshold for activation, particularly those caused by low-intensity stimuli. Consequently, central sensitization occurs [30]. The activation of group I mGluRs by glutamate also plays a significant role in the development of central sensitization. While these receptors are not involved in baseline nociception [177, 178], their activation is crucial for the activity-dependent central sensitization driven by C-fiber activity [179,180,181].
In addition to the upregulation of glutamate receptors in the development of central sensitization, impaired clearance of glutamate at the synapse between primary and secondary neurons in the brainstem has also been proposed. In healthy synapses, nearly all released glutamate in the synaptic cleft is taken up by astrocytes through excitatory amino acid transporters (EAATs) [73]. Within these astrocytes, glutamate is converted into glutamine, and then released in this non-toxic form to neurons, where it can be taken up and converted back into glutamate for repeated use [73]. In an animal model of chronic migraine, downregulation of EAAT2 transporters in the TNC has been observed [182]. This would limit the clearance of glutamate from the synaptic cleft, thereby causing overexcitation which could contribute to central sensitization. In contrast, up-regulation of these transporters has been shown to alleviate central sensitization in a rat model of chronic migraine [182].
If presynaptic neurons release a high amount of glutamate, and astrocytes fail to clear it from the synaptic cleft, it can lead to excitotoxicity. Excitotoxicity refers to repetitive neuronal excitation that causes neuronal damage and ultimately cell death [73]. In neurons, overactivation of glutamate receptors leads to excessive production of free radicals by mitochondria. Once the production of free radicals exceeds the cell’s antioxidant defense capacity, oxidative stress occurs [183]. On the other hand, nearby microglial cells, which express NMDA and mGlu receptors, can be directly triggered by glutamate to release inflammatory mediators [184], leading to neuroinflammation. This creates the neurotoxic triad of excitotoxicity, oxidative stress, and neuroinflammation—each capable of inducing and perpetuating the other two states. This triad has been proposed as an underlying mechanism implicated in neurological disorders, including migraine [185], which has also been associated with central sensitization (Fig. 3). One proposed mechanism involves the enhancement of NMDA receptor phosphorylation by reactive oxygen species (ROS), driving central sensitization [186, 187]. Phosphorylation of AMPA and NMDA receptors enhances their activity and density, resulting in postsynaptic hyperexcitability [188, 189]. Additionally, neuroinflammation, mediated by activated microglia, plays a significant role in the development of central sensitization. Proinflammatory cytokines and chemokines released by microglia affect both presynaptic and postsynaptic neurons [190]. Presynaptically, they enhance glutamate release, while postsynaptically, they positively modulate NMDA and AMPA receptors. For instance, interleukin-1 beta (IL-1β) increases excitatory transmission through NMDA receptor phosphorylation and reduces inhibitory signaling mediated by gamma-aminobutyric acid (GABA) and glycine [191]. Similarly, tumor necrosis factor alpha (TNF-α), also produced by microglia, binds to postsynaptic receptors in second-order neurons, amplifying NMDA and AMPA receptor responses [192, 193].
Central Sensitization Model Induced by the Neurotoxic Triad. (1) Excessive glutamate release and reduced reuptake of glutamate by astrocytes (through EAAT) leads to excitotoxicity, triggering massive Ca2+ influx in second-order neurons, mainly through NMDA receptors. (2) This eventually leads to the production of ROS by mitochondria. (3) ROS enhances NMDA receptor phosphorylation, driving central sensitization. (4) High levels of ROS surpass the cell’s antioxidant capacity causing oxidative stress in second-order neurons. (5) Available glutamate in the synaptic cleft can activate its NMDA and mGlu receptors on nearby microglial cells. (6) In response, microglial cells release inflammatory mediators that increase glutamate release presynaptically, enhance NMDA and AMPA receptor activity postsynaptically (through phosphorylation), and suppress inhibitory GABA and glycine signaling, further amplifying sensitization. These interconnected events form a triad of excitotoxicity, oxidative stress, and neuroinflammation, which not only potentiate central sensitization but also sustain it. NMDA: N-methyl-D-aspartic acid, AMPA: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, mGluRs: Metabotropic glutamate receptors, EAAT: Excitatory amino acid transporters, ROS: Reactive oxygen species. Ca2+: calcium, GABA: gamma-aminobutyric acid. Created in https://BioRender.com
CGRP’s role in central sensitization
CGRP participates in central sensitization through postsynaptic CGRP receptors. Activation of CGRP receptors in the spinal cord leads to mechanical hyperalgesia and increases the sensitivity of dorsal horn neurons in rats [31]. One potential mechanism for heightened sensitivity could be an increase in the number of CGRP receptors. In an animal study, injection of CGRP elevated sensitivity to mechanical stimuli in mice. Notably, this effect was even more pronounced in mice with increased expression of the CGRP receptor subunit [194]. Additionally, CGRP potentiates central sensitization through the activation of some kinase enzymes involved in triggering prolonged increases in gene transcription [195, 196]. CGRP has been found to enhance the release of SP and glutamate from rat spinal dorsal horn neurons during mechanical nociception [146]. These findings suggest that, although CGRP plays a central role in sensitization, part of its effects may be mediated indirectly through SP and glutamate.
While both glutamate and CGRP play key roles in central sensitization, their effects could be different in nature and dimension. In a study by Khodorova et al., they used subcutaneous injection of endothelin-1 (a potent vasoconstrictor) in rats in an in vivo model [197]. Subcutaneous injection of endothelin-1 into the hind paw of rodents results in local nociceptive sensitization or hyperalgesia [198, 199]. In this study, the initial rise in endothelin-1-induced mechanical allodynia, which remains unchanged by CGRP receptor antagonists, suggests that mediators released upon endothelin receptor activation may be involved in developing allodynia. They propose that glutamate, present in primary afferent neuron axons, and released in peripheral tissues following nerve stimulation, might also be released in the skin via neuronal endothelin receptor activation. This release of glutamate could subsequently stimulate a group of small-diameter, peptide-containing neurons, prompting the release of CGRP and possibly other peptides. Their findings indicate that blocking local NMDA receptors almost completely eliminates mechanical allodynia caused by endothelin-1. Interestingly, co-injection of NMDA receptor antagonists with endothelin-1 also prevented allodynia. In contrast, co-injection of the CGRP antagonist attenuated only the later phase of allodynia (> 30 min). This suggests that CGRP might play a role in maintaining, rather than initiating, the mechanisms of sensitization. However, it is important to note that evidence supporting the hyperalgesic effect of endothelin-1 and its subsequent mechanisms is derived from animal models, and its extrapolation to humans should be approached with caution [200].
Therapeutic potential and challenges in targeting glutamate and CGRP
NMDA receptor antagonists have been studied for their potential to treat and prevent migraine. This includes drugs like ketamine, which directly blocks the NMDA receptor; and medications like memantine, which is a partial antagonist to the NMDA receptor but with lower affinity than ketamine [201, 202]. A recent meta-analysis that included 38 clinical trials, compared memantine efficacy with the guideline-recommended prophylactic agents for migraine, including anti-CGRP monoclonal antibodies [202]. They showed that among all the interventions studied, memantine showed the second-largest decrease in migraine days and achieved the highest 50% response rate [202].
CNS side effects associated with glutamate antagonists have limited the enthusiasm for their use in migraine treatment [203]. Given that glutamate is a key neurotransmitter in the brain, altering its signaling is likely to result in CNS-related side effects. Modulating glutamate pathways can lead to adverse psychotomimetic effects such as hallucinations, delusions, paranoia, delirium, and impairments in learning, memory, and motor control [204]. Reported side effects of memantine (as a preventive option in migraine patients) include agitation, confusion, dizziness, weight loss, fatigue, rash, somnolence, weakness, anxiety, and depression [205, 206]. For ketamine, side effects such as dissociation, sedation, and blood pressure changes have been observed [207,208,209]. Frequent use of ketamine is linked to neurocognitive impairment, especially working and episodic memory deficits [210]. The very effects which have restricted ketamine’s clinical use, have also made it attractive to recreational users. Ketamine is highly addictive, being known on the street as ‘Special K’ [211]. On the other hand, to date, there have been no head-to-head studies of glutamate receptor antagonists with anti-CGRP medications, so most comparisons have been based on indirect estimates and have involved small sample sizes. Since glutamate is not only present endogenously in the brain, but is also found in the diet, with free forms of glutamate and aspartate (dietary excitotoxins that activate NMDA receptors) being commonly found as food additives [212], there is also increasing interest in the idea of dietary modulation as a potential treatment for migraine [213].
As stated above, there is great interest in the use of anti-CGRP monoclonal antibodies for migraine treatment. These work by blocking the CGRP pathway and represent the first preventive treatments specifically developed for migraine, with generally favorable safety profiles [214,215,216,217,218]. Later studies have validated their effectiveness, even in patients who were resistant to other treatments [219,220,221].
Based on published data so far, around one-third of patients show a strong response to anti-CGRP monoclonal antibodies, while another third does not respond at all [8,9,10,11]. Some potential explanations exist for the non-responders to anti-CGRP medication. The first pertains to chronic migraineurs whose attacks are predominantly driven by central mechanisms, rendering anti-CGRP medications less effective. The second explanation is rooted in anatomical evidence indicating a lack of co-localization between CGRP and its receptors [105]. Specifically, C fibers contain CGRP, while Aδ fibers house CGRP receptors. For example, studies show that fremanezumab, a CGRP antagonist, inhibits Aδ fibers, but not C fibers [222]. At the peripheral level, either in episodic or chronic migraine, antagonizing CGRP or its receptors may inhibit Aδ fiber activation. However, C fibers can still be activated through other mechanisms, such as cortical spreading depression and inflammation [222, 223].
Some limitations of anti-CGRP medications include their high cost and a lack of long-term data from randomized trials. Notably, these newly developed medications are still unavailable in many countries. In fact, a recent survey by the international headache society across 84 countries reported that new migraine treatments, such as rimegepant and erenumab (both CGRP receptor antagonists), remain largely inaccessible across Latin America, Asia, and Africa [224]. As a result, these treatments were initially recommended as second or third-line options for chronic migraine, though more recent guidelines have endorsed them as first-line therapies [225, 226].
Notably, the issue extends beyond non-responders. While this family of medications is generally safe, some patients experience unavoidable side effects. These impact multiple organs and systems, including the gastrointestinal, cardiovascular, nervous, respiratory, immune, and musculoskeletal systems, as well as issues with the skin and hair. Some examples of these side effects include constipation, hypertension, palpitations, anxiety, vertigo/dizziness, paresthesia, insomnia, flu-like symptoms, hypersensitivity reactions, myalgia, and alopecia [227,228,229,230]. Such adverse effects can diminish patients’ enthusiasm for this treatment. Moreover, since these medications are newly developed, limited data is available about their long-term safety. A recent observational study revealed that during a 1.5-year follow-up, 1.6% of patients receiving anti-CGRP medications experienced moderate to severe cardiovascular events despite having no cardiovascular risk factors or hypertension at baseline [231]. These medications are also contraindicated during pregnancy and lactation, as well as in individuals with vascular disorders or ischemia [12,13,14,15,16], which limits their use in certain populations.
Clinical implications: the role of external triggers
Extensive reports exist regarding the role of triggers in migraine [232,233,234]. It is important to note here that a reported migraine trigger may sometimes be confused with symptoms of the premonitory phase, which is already part of the migraine attack process [235]. Based on this notion, it has been suggested that studies investigating “migraine” and its “triggers” should carefully assess the specific stages of the migraine cycle, considering not only the end of the previous attack, but also the onset of the next one [235]. In addition to this important caveat, it is also important that future research examine the potential mechanisms for how a specific exposure may affect key aspects of migraine, such as effects on CGRP, glutamate, and vasodilation. One common argument against certain substances having the ability to affect migraine is that the BBB protects the brain from chemicals in the blood. Two key considerations challenge this. First, as mentioned earlier, not everyone has an intact BBB capable of fully shielding against toxins and triggers. Although evidence remains inconclusive regarding BBB permeability in migraine patients, it is suggested that the barrier may become transiently permeable during an attack [236]. Second, and crucially, if migraine pain begins with the activation of primary afferents in the dura, we must acknowledge that the TG and dura are outside the BBB’s protection. Therefore, these critical regions for the formation of pain signals are vulnerable to external triggers circulating in the blood. This provides a great opportunity to develop targeted prevention and intervention strategies. In this regard, a non-pharmaceutical approach that controls the peripheral level of glutamate could be a valuable alternative option for preventing attacks.
Monosodium glutamate (MSG) is recognized as the third most common dietary trigger for headaches [237,238,239,240]. Studies have shown that consuming 150 mg/kg of MSG can lead to increased reports of headache, nausea, and vomiting [237,238,239,240] - symptoms commonly linked to acute migraine attacks. In individuals with fibromyalgia, lowering dietary MSG intake has also been associated with reduced musculoskeletal pain and reduced headache occurrence [241]. Using a glutamate-selective biosensor, glutamate levels were measured in the TG of an anesthetized male rat before and after a dose of MSG [242]. At time zero, MSG was injected into the carotid artery, resulting in a sustained rise in glutamate levels in the TG. Then, these afferents activated second-order neurons in the brain stem nuclei, and, finally, higher areas involved in pain perception of the brain. This aligns with earlier observations that MSG, when applied peripherally, enhances the ongoing spiking activity in brainstem neurons [25]. As free glutamate (not-bound to a protein) is found as a naturally occurring amino acid in some foods, and is also widely used in the food industry as a flavor-enhancing food additive, diet serves as an important source of exogenous glutamate in blood [212, 243, 244].
Interestingly, diet may also play an important protective role against migraine, as magnesium and several other nutrients have shown efficacy in combating glutamate excitotoxicity, inflammation, and oxidative stress, with promising results in clinical trials involving migraine patients (as detailed in our previous review [213]). Additionally, some studies indicate that specific dietary factors may also influence circulating CGRP levels, with a potential impact on migraine outcomes (reviewed in [245]). Therefore, future research is needed to fully understand how diet may be influencing migraine through effects on glutamate and CGRP, and whether dietary interventions targeting these neuro-messengers may have anti-migraine effects.
What have we learned about CGRP-glutamate dynamics, and what remains unknown?
As discussed in this review, migraine originates peripherally and progresses centrally, reaching a point in chronic migraine where it may become entirely centralized. Anti-CGRP therapies primarily target peripheral pain mechanisms, such as neurogenic inflammation and vasodilation, but central mechanisms also play a significant role in migraine. In some patients, central mechanisms may be more dominant than peripheral ones, meaning that targeting CGRP alone may not be enough to address the central components of their migraines, such as glutamate-mediated excitotoxicity. This hypothesis is backed up by evidence showing that the presence of central sensitization and its associated symptoms were particularly predictive of treatment failure. In this regard, allodynia is predictive of a poor response to anti-CGRP medications [246]. Since allodynia arises from central sensitization, these treatments may not be sufficient to reverse chronic migraine once central sensitization is established. The central role of glutamate in higher regions of the pain pathways is well-supported [81, 82], making it logical to hypothesize that certain patients may benefit more from targeting glutamate. This may also apply to migraineurs with aura. Cortical spreading depression, the neural phenomenon underlying aura, occurs within the brain itself, indicating that aura symptoms originate from the central nervous system. The involvement of glutamate in mediating cortical spreading depression through NMDA receptors is widely supported [247, 248]. Therefore, it is reasonable to hypothesize that this migraine subtype may be particularly responsive to glutamate-targeted interventions.
The mechanisms discussed in this paper highlight the complexity of migraine and the variability in its clinical presentation across patients. Each mechanism may contribute to migraine pathophysiology and should not be considered in isolation. Instead, a comprehensive view that integrates these diverse processes is needed. Accordingly, the development of varied treatment strategies is essential. Future research should focus on therapies that target both peripheral and central components to more effectively address the full spectrum of migraine pathophysiology.
Conclusion
The evidence supports a model in which glutamate plays an upstream role in stimulating CGRP release, while CGRP, in turn, enhances glutamate signaling, creating a feedback loop that may contribute to sustained pain in migraine. With growing recognition of glutamate’s role in both peripheral and central sensitization, targeting glutamate pathways presents a promising therapeutic avenue—particularly for individuals who do not respond to CGRP-based treatments. On the other hand, given the involvement of multiple underlying mechanisms and the limitations and challenges associated with current therapies, it is logical to adopt diverse approaches to effectively treat migraine. Furthermore, as evidence continues to support migraine initiation at the dura and TG, future research should explore strategies to reduce peripheral glutamate exposure.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- CGRP:
-
Calcitonin gene-related peptide
- BBB:
-
Blood-brain barrier
- TG:
-
Trigeminal ganglion
- TNC:
-
Trigeminal nucleus caudalis
- TCC:
-
Trigeminal cervical complex
- CNS:
-
Central nervous system
- iGluRs:
-
Ionotropic glutamate receptors
- mGluRs:
-
Metabotropic glutamate receptors
- NMDA:
-
N-methyl-D-aspartic acid
- AMPA:
-
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
- Ca2+ :
-
Calcium
- NO:
-
Nitric oxide
- cGMP:
-
Cyclic guanosine monophosphate
- cAMP:
-
Cyclic adenosine monophosphate
- MSG:
-
Monosodium glutamate
- SP:
-
Substance P
- Mg2+ :
-
Magnesium
- EAATs:
-
Excitatory amino acid transporters
- ROS:
-
Reactive oxygen species
- IL-1β:
-
Interleukin-1 beta
- GABA:
-
Gamma-aminobutyric acid
- TNF-α:
-
Tumor necrosis factor-alpha
References
Burstein R, Noseda R, Borsook D (2015) Migraine: multiple processes, complex pathophysiology. J Neurosci 35(17):6619–6629
Noseda R, Burstein R (2013) Migraine pathophysiology: anatomy of the trigeminovascular pathway and associated neurological symptoms, cortical spreading depression, sensitization, and modulation of pain. Pain 154:S44–S53
Goadsby PJ, Edvinsson L (1994) Human in vivo evidence for trigeminovascular activation in cluster headache neuropeptide changes and effects of acute attacks therapies. Brain 117(3):427–434
Asghar MS, Becerra L, Larsson HB, Borsook D, Ashina M (2016) Calcitonin gene-related peptide modulates heat nociception in the human brain-an fMRI study in healthy volunteers. PLoS ONE 11(3):e0150334
Lambru G, Hill B, Murphy M, Tylova I, Andreou AP (2020) A prospective real-world analysis of erenumab in refractory chronic migraine. J Headache Pain.;21(1)
Andreou AP, Fuccaro M, Hill B, Murphy M, Caponnetto V, Kilner R et al (2022) Two-year effectiveness of erenumab in resistant chronic migraine: a prospective real-world analysis. J Headache Pain.;23(1)
Alasad YW, Asha MZ (2020) Monoclonal antibodies as a preventive therapy for migraine: a meta-analysis. Clin Neurol Neurosurg 195:105900
Lee MJ, Al-Karagholi MA-M, Reuter U (2023) New migraine prophylactic drugs: current evidence and practical suggestions for non-responders to prior therapy. Cephalalgia 43(2):03331024221146315
Raffaelli B, Fitzek M, Overeem LH, Storch E, Terhart M, Reuter U (2023) Clinical evaluation of super-responders vs. non-responders to CGRP (-receptor) monoclonal antibodies: a real-world experience. J Headache Pain 24(1):16
Ziegeler C, May A (2019) Non-responders to treatment with antibodies to the CGRP-receptor May profit from a switch of antibody class. Headache 60(2):469–470
Burgalassi A, Romozzi M, Vigani G, De Icco R, Raffaelli B, Boccalini A et al (2024) Outcomes, unmet needs, and challenges in the management of patients who withdraw from anti-CGRP monoclonal antibodies: A prospective cohort study. Cephalalgia 44(11):03331024241273968
Nguyen JL, Munshi K, Peasah SK, Swart EC, Kohli M, Henderson R et al (2022) Trends in utilization and costs of migraine medications, 2017–2020. J Headache Pain 23(1):111
Haanes KA, Edvinsson L, Sams A (2020) Understanding side-effects of anti-CGRP and anti-CGRP receptor antibodies. J Headache Pain 21:1–2
Bussiere JL, Davies R, Dean C, Xu C, Kim KH, Vargas HM et al (2019) Nonclinical safety evaluation of erenumab, a CGRP receptor inhibitor for the prevention of migraine. Regul Toxicol Pharmacol 106:224–238
Gangula P, Dong Y, Wimalawansa S, Yallampalli C (2002) Infusion of pregnant rats with calcitonin gene-related peptide (CGRP) 8–37, a CGRP receptor antagonist, increases blood pressure and fetal mortality and decreases fetal growth. Biol Reprod 67(2):624–629
MaassenVanDenBrink A, Meijer J, Villalón CM, Ferrari MD (2016) Wiping out CGRP: potential cardiovascular risks. Trends Pharmacol Sci 37(9):779–788
Johnson KW, Morin SM, Wroblewski VJ, Johnson MP (2019) Peripheral and central nervous system distribution of the CGRP neutralizing antibody [125I] galcanezumab in male rats. Cephalalgia 39(10):1241–1248
Noseda R, Schain AJ, Melo-Carrillo A, Tien J, Stratton J, Mai F et al (2020) Fluorescently-labeled fremanezumab is distributed to sensory and autonomic ganglia and the dura but not to the brain of rats with uncompromised blood brain barrier. Cephalalgia 40(3):229–240
Karsan N, Goadsby PJ (2018) Biological insights from the premonitory symptoms of migraine. Nat Reviews Neurol 14(12):699–710
Basedau H, Sturm L-M, Mehnert J, Peng K-P, Schellong M, May A (2022) Migraine monoclonal antibodies against CGRP change brain activity depending on ligand or receptor target–an fMRI study. Elife 11:e77146
Rorabaugh J, Hellriegel E, Ghibellini G, Zeng H, Lu X (2023) CNS target engagement by the anti-CGRP mAb fremanezumab in the cerebrospinal fluid of healthy volunteers. Cephalalgia 43(1S):1–2
Levy D, Burstein R, Strassman AM (2005) Calcitonin gene–related peptide does not excite or sensitize meningeal nociceptors: implications for the pathophysiology of migraine. Annals Neurology: Official J Am Neurol Association Child Neurol Soc 58(5):698–705
Watanabe M, Mishina M, Inoue Y (1994) Distinct gene expression of the N-methyl-D-aspartate receptor channel subunit in peripheral neurons of the mouse sensory ganglia and adrenal gland. Neurosci Lett 165(1–2):183–186
Tallaksen-Greene SJ, Young AB, Penney JB, Beitz AJ (1992) Excitatory amino acid binding sites in the trigeminal principal sensory and spinal trigeminal nuclei of the rat. Neurosci Lett 141(1):79–83
O’Brien M, Cairns BE (2016) Monosodium glutamate alters the response properties of rat trigeminovascular neurons through activation of peripheral NMDA receptors. Neuroscience 334:236–244
Crivellaro G, Tottene A, Vitale M, Melone M, Casari G, Conti F et al (2021) Specific activation of GluN1-N2B NMDA receptors underlies facilitation of cortical spreading depression in a genetic mouse model of migraine with reduced astrocytic glutamate clearance. Neurobiol Dis 156:105419
Ferrari MD, Odink J, Bos K, Malessy M, Bruyn G (1990) Neuroexcitatory plasma amino acids are elevated in migraine. Neurology 40(10):1582
Martínez F, Castillo J, Rodríguez JR, Leira R, Noya M (1993) Neuroexcitatory amino acid levels in plasma and cerebrospinal fluid during migraine attacks. Cephalalgia 13(2):89–93
Peek AL, Rebbeck T, Puts NA, Watson J, Aguila M-ER, Leaver AM (2020) Brain GABA and glutamate levels across pain conditions: a systematic literature review and meta-analysis of 1H-MRS studies using the MRS-Q quality assessment tool. NeuroImage 210:116532
Sarchielli P, Di Filippo M, Nardi K, Calabresi P (2007) Sensitization, glutamate, and the link between migraine and fibromyalgia. Curr Pain Headache Rep 11:343–351
Sun R-Q, Tu Y-J, Lawand NB, Yan J-Y, Lin Q, Willis WD (2004) Calcitonin gene-related peptide receptor activation produces PKA-and PKC-dependent mechanical hyperalgesia and central sensitization. J Neurophysiol 92(5):2859–2866
Sun R-Q, Lawand NB, Willis WD (2003) The role of calcitonin gene-related peptide (CGRP) in the generation and maintenance of mechanical allodynia and hyperalgesia in rats after intradermal injection of capsaicin. Pain 104(1–2):201–208
Benbow T, Ekbatan MR, Wang GHY, Teja F, Exposto FG, Svensson P et al (2022) Systemic administration of monosodium glutamate induces sexually dimorphic headache-and nausea-like behaviours in rats. Pain 163(9):1838–1853
Kageneck C, Nixdorf-Bergweiler BE, Messlinger K, Fischer MJ (2014) Release of CGRP from mouse brainstem slices indicates central inhibitory effect of triptans and kynurenate. J Headache Pain 15:1–9
Gazerani P, Au S, Dong X, Kumar U, Arendt-Nielsen L, Cairns BE (2010) Botulinum neurotoxin type A (BoNTA) decreases the mechanical sensitivity of nociceptors and inhibits neurogenic vasodilation in a craniofacial muscle targeted for migraine prophylaxis. PAIN® 151(3):606–616
Tringali G, Currò D, Navarra P (2018) Perampanel inhibits calcitonin gene-related peptide release from rat brainstem in vitro. J Headache Pain 19(1):107
Buzzi M, Tassorelli C, Nappi G (2003) Peripheral and central activation of trigeminal pain pathways in migraine: data from experimental animal models. Cephalalgia 23(1suppl):1–4
Oshinsky ML, Luo J (2006) Neurochemistry of trigeminal activation in an animal model of migraine. Headache: J Head Face Pain 46:S39–S44
Oshinsky ML, Gomonchareonsiri S (2007) Episodic dural stimulation in awake rats: a model for recurrent headache. Headache: J Head Face Pain 47(7):1026–1036
Iyengar S, Johnson KW, Ossipov MH, Aurora SK (2019) CGRP and the trigeminal system in migraine. Headache: J Head Face Pain 59(5):659–681
Ashina M, Hansen JM, Do TP, Melo-Carrillo A, Burstein R, Moskowitz MA (2019) Migraine and the trigeminovascular system—40 years and counting. Lancet Neurol 18(8):795–804
Edvinsson L, Haanes KA, Warfvinge K, Krause DN (2018) CGRP as the target of new migraine therapies — successful translation from bench to clinic. Nat Reviews Neurol 14(6):338–350
Liu Y, Broman J, Edvinsson L (2004) Central projections of sensory innervation of the rat superior sagittal sinus. Neuroscience 129(2):431–437
Basbaum AI, Bautista DM, Scherrer G, Julius D (2009) Cellular and molecular mechanisms of pain. Cell 139(2):267–284
Patel NM, Jozsa F, Das JM (2022) Neuroanatomy, spinal trigeminal nucleus. StatPearls Publishing, StatPearls [Internet]
Woolfall P, Coulthard A (2001) Trigeminal nerve: anatomy and pathology. Br J Radiol 74(881):458–467
Andres KH, von Düring M, Muszynski K, Schmidt RF (1987) Nerve fibres and their terminals of the dura mater encephali of the rat. Anat Embryol 175(3):289–301
Messlinger K, Dostrovsky JO, Strassman AM (2006) Anatomy and physiology of head pain. Lippincott Williams & Wilkins, Philadelphia;, The headaches, pp 95–109
Keller JT, Saunders MC, Beduk A, Jollis JG (1985) Innervation of the posterior fossa dura of the Cat. Brain Res Bull 14(1):97–102
Uddman R, Edvinsson L, Ekman R, Kingman T, McCulloch J (1985) Innervation of the feline cerebral vasculature by nerve fibers containing calcitonin gene-related peptide: trigeminal origin and co-existence with substance P. Neurosci Lett 62(1):131–136
Davis K, Dostrovsky J (1988) Responses of feline trigeminal spinal tract nucleus neurons to stimulation of the middle meningeal artery and sagittal sinus. J Neurophysiol 59(2):648–666
Busch V, Frese A, Bartsch T (2004) The trigemino-cervical complex: integration of peripheral and central pain mechanisms in primary headache syndromes. Der Schmerz 18:404–410
Davis KD, Dostrovsky JO (1986) Activation of trigeminal brain-stem nociceptive neurons by dural artery stimulation. Pain 25(3):395–401
Bartsch T, Goadsby PJ (2003) Increased responses in trigeminocervical nociceptive neurons to cervical input after stimulation of the dura mater. Brain 126(8):1801–1813
GOADSBY PJ, ZAGAMI AS, STIMULATION OF THE SUPERIOR SAGITTAL SINUS INCREASES METABOLIC ACTIVITY, AND BLOOD FLOW IN CERTAIN REGIONS OF THE BRAINSTEM AND UPPER CERVICAL SPINAL CORD OF THE CAT (1991) Brain 114(2):1001–1011
Malick A, Strassman RM, Burstein R (2000) Trigeminohypothalamic and reticulohypothalamic tract neurons in the upper cervical spinal cord and caudal medulla of the rat. J Neurophysiol 84(4):2078–2112
Noseda R, Jakubowski M, Kainz V, Borsook D, Burstein R (2011) Cortical projections of functionally identified thalamic trigeminovascular neurons: implications for migraine headache and its associated symptoms. J Neurosci 31(40):14204–14217
Andreou A, Trimboli M, Al-Kaisy A, Murphy M, Palmisani S, Fenech C et al (2018) Prospective real‐world analysis of OnabotulinumtoxinA in chronic migraine post‐National Institute for health and care excellence UK technology appraisal. Eur J Neurol 25(8):1069–e83
Dodick DW (2019) CGRP ligand and receptor monoclonal antibodies for migraine prevention: evidence review and clinical implications. Cephalalgia 39(3):445–458
Neurol L, Pain JH (2019) Complicated decisions on new migraine-prevention therapies. Lancet Neurol 18:5–6
Lundblad C, Haanes KA, Grände G, Edvinsson L (2015) Experimental inflammation following dural application of complete Freund’s adjuvant or inflammatory soup does not alter brain and trigeminal microvascular passage. J Headache Pain 16:1–9
Messlinger K, Russo AF (2019) Current Understanding of trigeminal ganglion structure and function in headache. Cephalalgia 39(13):1661–1674
Eftekhari S, Gaspar RC, Roberts R, Chen T-B, Zeng Z, Villarreal S et al (2016) Localization of CGRP receptor components and receptor binding sites in rhesus monkey brainstem: A detailed study using in situ hybridization, Immunofluorescence, and autoradiography. J Comp Neurol 524(1):90–118
Riedel G, Platt B, Micheau J (2003) Glutamate receptor function in learning and memory. Behav Brain Res 140(1–2):1–47
Ohgi Y, Futamura T, Hashimoto K (2015) Glutamate signaling in synaptogenesis and NMDA receptors as potential therapeutic targets for psychiatric disorders. Curr Mol Med 15(3):206–221
Plitman E, Nakajima S, de la Fuente-Sandoval C, Gerretsen P, Chakravarty MM, Kobylianskii J et al (2014) Glutamate-mediated excitotoxicity in schizophrenia: a review. Eur Neuropsychopharmacol 24(10):1591–1605
Dong X-x, Wang Y, Qin Z-h (2009) Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol Sin 30(4):379–387
Pitt D, Werner P, Raine CS (2000) Glutamate excitotoxicity in a model of multiple sclerosis. Nat Med 6(1):67–70
Olney J, Collins R, Sloviter R (1986) Excitotoxic mechanisms of epileptic brain damage. Adv Neurol 44:857–877
Petrenko AB, Shimoji K (2001) A possible role for glutamate receptor-mediated excitotoxicity in chronic pain. J Anesth 15(1):39–48
Longoni M, Ferrarese C (2006) Inflammation and excitotoxicity: role in migraine pathogenesis. Neurol Sci 27:s107–s10
El Mestikawy S, Wallén-Mackenzie Å, Fortin GM, Descarries L, Trudeau L-E (2011) From glutamate co-release to vesicular synergy: vesicular glutamate transporters. Nat Rev Neurosci 12(4):204–216
McKenna MC (2007) The glutamate-glutamine cycle is not stoichiometric: fates of glutamate in brain. J Neurosci Res 85(15):3347–3358
Willard SS, Koochekpour S (2013) Glutamate, glutamate receptors, and downstream signaling pathways. Int J Biol Sci 9(9):948
Fernández-Montoya J, Avendaño C, Negredo P (2018) The glutamatergic system in primary somatosensory neurons and its involvement in sensory Input-Dependent plasticity. Int J Mol Sci 19(1):69
Quartu M, Serra MP, Ambu R, Lai ML, Del Fiacco M (2002) AMPA-type glutamate receptor subunits 2/3 in the human trigeminal sensory ganglion and subnucleus caudalis from prenatal ages to adulthood. Mech Ageing Dev 123(5):463–471
Sahara Y, Noro N, Iida Y, Soma K, Nakamura Y (1997) Glutamate receptor subunits GluR5 and KA-2 are coexpressed in rat trigeminal ganglion neurons. J Neurosci 17(17):6611–6620
Furuyama T, Kiyama H, Sato K, Park HT, Maeno H, Takagi H et al (1993) Region-specific expression of subunits of ionotropic glutamate receptors (AMPA-type, KA-type and NMDA receptors) in the rat spinal cord with special reference to nociception. Mol Brain Res 18(1–2):141–151
Huettner JE (1990) Glutamate receptor channels in rat DRG neurons: activation by Kainate and quisqualate and Blockade of desensitization by Con A. Neuron 5(3):255–266
Sharp JW, Sagar SM, Hisanaga K, Jasper P, Sharp FR (1990) The NMDA receptor mediates cortical induction of Fos and Fos-related antigens following cortical injury. Exp Neurol 109(3):323–332
Greenamyre JT, Young AB, Penney JB (1984) Quantitative autoradiographic distribution of L-[3H] glutamate-binding sites in rat central nervous system. J Neurosci 4(8):2133–2144
Halpain S, Wieczorek C, Rainbow TC (1984) Localization of L-glutamate receptors in rat brain by quantitative autoradiography. J Neurosci 4(9):2247–2258
Liu H, Mantyh PW, Basbaum AI (1997) NMDA-receptor regulation of substance P release from primary afferent nociceptors. Nature 386(6626):721–724
Bardoni R (2013) Role of presynaptic glutamate receptors in pain transmission at the spinal cord level. Curr Neuropharmacol 11(5):477–483
Bereiter DA, Benetti AP (1996) Excitatory amino release within spinal trigeminal nucleus after mustard oil injection into the temporomandibular joint region of the rat. Pain 67(2):451–459
Ferrari MD, Klever RR, Terwindt GM, Ayata C, van den Maagdenberg AM (2015) Migraine pathophysiology: lessons from mouse models and human genetics. Lancet Neurol 14(1):65–80
Gazerani P, Dong X, Wang M, Kumar U, Cairns BE (2010) Sensitization of rat facial cutaneous mechanoreceptors by activation of peripheral N-methyl-d-aspartate receptors. Brain Res 1319:70–82
Lam DK, Sessle BJ, Hu JW (2009) Glutamate and capsaicin effects on trigeminal nociception I: activation and peripheral sensitization of deep craniofacial nociceptive afferents. Brain Res 1251:130–139
Cairns BE, Svensson P, Wang K, Hupfeld S, Graven-Nielsen T, Sessle BJ et al (2003) Activation of peripheral NMDA receptors contributes to human pain and rat afferent discharges evoked by injection of glutamate into the masseter muscle. J Neurophysiol 90(4):2098–2105
Lee J, Saloman JL, Weiland G, Auh Q-S, Chung M-K, Ro JY (2012) Functional interactions between NMDA receptors and TRPV1 in trigeminal sensory neurons mediate mechanical hyperalgesia in the rat masseter muscle. PAIN® 153(7):1514–1524
Guerrero-Toro C, Koroleva K, Ermakova E, Gafurov O, Abushik P, Tavi P et al (2022) Testing the role of glutamate NMDA receptors in peripheral trigeminal nociception implicated in migraine pain. Int J Mol Sci 23(3):1529
Alam Z, Coombes N, Waring RH, Williams AC, Steventon GB (1998) Plasma levels of neuroexcitatory amino acids in patients with migraine or tension headache. J Neurol Sci 156(1):102–106
Peres M, Zukerman E, Soares CS, Alonso E, Santos B, Faulhaber M (2004) Cerebrospinal fluid glutamate levels in chronic migraine. Cephalalgia 24(9):735–739
Ferrari A, Spaccalopelo L, Pinetti D, Tacchi R, Bertolini A (2009) Effective prophylactic treatments of migraine lower plasma glutamate levels. Cephalalgia 29(4):423–429
Garthwaite J, Charles SL, Chess-Williams R (1988) Endothelium-derived relaxing factor release on activation of NMDA receptors suggests role as intercellular messenger in the brain. Nature 336(6197):385–388
Christopherson KS, Hillier BJ, Lim WA, Bredt DS (1999) PSD-95 assembles a ternary complex with theN-Methyl-D-aspartic acid receptor and a bivalent neuronal NO synthase PDZ domain. J Biol Chem 274(39):27467–27473
LeMaistre JL, Sanders SA, Stobart MJ, Lu L, Knox JD, Anderson HD et al (2012) Coactivation of NMDA receptors by glutamate and-serine induces dilation of isolated middle cerebral arteries. J Cereb Blood Flow Metabolism 32(3):537
Storer RJ, Akerman S, Goadsby PJ (2004) Calcitonin gene-related peptide (CGRP) modulates nociceptive trigeminovascular transmission in the Cat. Br J Pharmacol 142(7):1171–1181
Schou WS, Ashina S, Amin FM, Goadsby PJ, Ashina M (2017) Calcitonin gene-related peptide and pain: a systematic review. J Headache Pain 18:1–17
Messlinger K, Lennerz JK, Eberhardt M, Fischer MJ (2012) CGRP and NO in the trigeminal system: mechanisms and role in headache generation. Headache: J Head Face Pain 52(9):1411–1427
Hay DL, Walker CS (2017) CGRP and its receptors. Headache: J Head Face Pain 57(4):625–636
Eftekhari S, Salvatore CA, Johansson S, Chen T-b, Zeng Z, Edvinsson L (2015) Localization of CGRP, CGRP receptor, PACAP and glutamate in trigeminal ganglion. Relation to the blood–brain barrier. Brain Res 1600:93–109
Warfvinge K, Edvinsson L (2019) Distribution of CGRP and CGRP receptor components in the rat brain. Cephalalgia 39(3):342–353
Edvinsson L, Warfvinge K (2017) Recognizing the role of CGRP and CGRP receptors in migraine and its treatment. Cephalalgia 39(3):366–373
Eftekhari S, Warfvinge K, Blixt FW, Edvinsson L (2013) Differentiation of nerve fibers storing CGRP and CGRP receptors in the peripheral trigeminovascular system. J Pain 14(11):1289–1303
Andres K, Von Düring M, Muszynski K, Schmidt R (1987) Nerve fibres and their terminals of the dura mater encephali of the rat. Anat Embryol 175:289–301
Strassman AM, Levy D (2006) Response properties of dural nociceptors in relation to headache. J Neurophysiol 95(3):1298–1306
Ebersberger A, Averbeck B, Messlinger K, Reeh P (1999) Release of substance P, calcitonin gene-related peptide and prostaglandin E2 from rat dura mater encephali following electrical and chemical stimulation in vitro. Neuroscience 89(3):901–907
Levy D, Strassman A (2002) Distinct sensitizing effects of the cAMP-PKA second messenger cascade on rat dural mechanonociceptors. J Physiol 538(2):483–493
Lennerz JK, Rühle V, Ceppa EP, Neuhuber WL, Bunnett NW, Grady EF et al (2008) Calcitonin receptor-like receptor (CLR), receptor activity‐modifying protein 1 (RAMP1), and calcitonin gene‐related peptide (CGRP) immunoreactivity in the rat trigeminovascular system: differences between peripheral and central CGRP receptor distribution. J Comp Neurol 507(3):1277–1299
Avona A, Burgos-Vega C, Burton MD, Akopian AN, Price TJ, Dussor G (2019) Dural calcitonin gene-related peptide produces female-specific responses in rodent migraine models. J Neurosci 39(22):4323–4331
Paige C, Plasencia-Fernandez I, Kume M, Papalampropoulou-Tsiridou M, Lorenzo L-E, David ET et al (2022) A female-specific role for calcitonin gene-related peptide (CGRP) in rodent pain models. J Neurosci 42(10):1930–1944
Covasala O, Stirn SL, Albrecht S, De Col R, Messlinger K (2012) Calcitonin gene-related peptide receptors in rat trigeminal ganglion do not control spinal trigeminal activity. J Neurophysiol 108(2):431–440
Fischer MJM, Koulchitsky S, Messlinger K (2005) The nonpeptide calcitonin gene-related peptide receptor antagonist BIBN4096BS lowers the activity of neurons with meningeal input in the rat spinal trigeminal nucleus. J Neurosci 25(25):5877–5883
Pozo-Rosich P, Storer R, Charbit A, Goadsby P (2015) Periaqueductal Gray calcitonin gene-related peptide modulates trigeminovascular neurons. Cephalalgia 35(14):1298–1307
Menon S, Buteri J, Roy B, Murrell M, Quinlan S, Macmillan J et al (2011) Association study of calcitonin gene-related polypeptide-alpha (CALCA) gene polymorphism with migraine. Brain Res 1378:119–124
Genome-wide association (2010) Study of migraine implicates a common susceptibility variant on 8q22. 1. Nat Genet 42(10):869–873
Iversen HK, Olesen J, Tfelt-Hansen P (1989) Intravenous nitroglycerin as an experimental model of vascular headache. Basic characteristics. Pain 38(1):17–24
Schytz HW, Birk S, Wienecke T, Kruuse C, Olesen J, Ashina M (2009) PACAP38 induces migraine-like attacks in patients with migraine without aura. Brain 132(1):16–25
Christiansen I, Thomsen L, Daugaard D, Ulrich V, Olesen J (1999) Glyceryl trinitrate induces attacks of migraine without aura in sufferers of migraine with aura. Cephalalgia 19(7):660–667
Lassen LH, Thomsen LL, Olesen J (1995) Histamine induces migraine via the H1-receptor. Support for the NO hypothesis of migraine. NeuroReport 6(11):1475–1479
Rahmann A, Wienecke T, Hansen J, Fahrenkrug J, Olesen J, Ashina M (2008) Vasoactive intestinal peptide causes marked cephalic vasodilation, but does not induce migraine. Cephalalgia 28(3):226–236
Pellesi L, Al-Karagholi MA-M, Chaudhry BA, Lopez CL, Snellman J, Hannibal J et al (2020) Two-hour infusion of vasoactive intestinal polypeptide induces delayed headache and extracranial vasodilation in healthy volunteers. Cephalalgia 40(11):1212–1223
Pellesi L, Al-Karagholi MA-M, De Icco R, Coskun H, Elbahi FA, Lopez-Lopez C et al (2021) Effect of vasoactive intestinal polypeptide on development of migraine headaches: a randomized clinical trial. JAMA Netw Open 4(8):e2118543–e
Schoonman GG, van der Grond J, Kortmann C, van der Geest RJ, Terwindt GM, Ferrari MD (2008) Migraine headache is not associated with cerebral or meningeal vasodilatation—a 3T magnetic resonance angiography study. Brain 131(8):2192–2200
Amin FM, Asghar MS, Hougaard A, Hansen AE, Larsen VA, de Koning PJ et al (2013) Magnetic resonance angiography of intracranial and extracranial arteries in patients with spontaneous migraine without aura: a cross-sectional study. Lancet Neurol 12(5):454–461
Lassen LH, Jacobsen VB, Haderslev PA, Sperling B, Iversen HK, Olesen J et al (2008) Involvement of calcitonin gene-related peptide in migraine: regional cerebral blood flow and blood flow velocity in migraine patients. J Headache Pain 9(3):151–157
Levy D, Strassman AM (2002) Mechanical response properties of A and C primary afferent neurons innervating the rat intracranial dura. J Neurophysiol 88(6):3021–3031
Ashina M, Migraine (2020) N Engl J Med 383(19):1866–1876
Hawkins RA (2009) The blood-brain barrier and glutamate. Am J Clin Nutr 90(3):867S–74S
Price MT, Olney JW, Lowry OH, Buchsbaum S (1981) Uptake of exogenous glutamate and aspartate by circumventricular organs but not other regions of brain. J Neurochem 36(5):1774–1780
Toklu HZ, Tümer N (2015) Oxidative stress, brain edema, blood–brain barrier permeability, and autonomic dysfunction from traumatic brain injury. Brain neurotrauma: molecular, neuropsychological, and rehabilitation aspects
Galea I (2021) The blood–brain barrier in systemic infection and inflammation. Cell Mol Immunol 18(11):2489–2501
Lochhead JJ, McCaffrey G, Quigley CE, Finch J, DeMarco KM, Nametz N et al (2010) Oxidative stress increases blood–brain barrier permeability and induces alterations in occludin during hypoxia–reoxygenation. J Cereb Blood Flow Metabolism 30(9):1625–1636
Gao X, Bayraktutan U (2023) Substance P reversibly compromises the integrity and function of blood-brain barrier. Peptides 167:171048
Abdel-Rahman A, Shetty A, Abou-Donia M (2002) Acute exposure to sarin increases blood brain barrier permeability and induces neuropathological changes in the rat brain: dose–response relationships. Neuroscience 113(3):721–741
Vazana U, Veksler R, Pell GS, Prager O, Fassler M, Chassidim Y et al (2016) Glutamate-mediated blood–brain barrier opening: implications for neuroprotection and drug delivery. J Neurosci 36(29):7727–7739
Kangrga I, Larew J, Randic M (1990) The effects of substance P and calcitonin gene-related peptide on the efflux of endogenous glutamate and aspartate from the rat spinal dorsal Horn in vitro. Neurosci Lett 108(1–2):155–160
Takhshid MA, Owji AA, Panjehshahin MR (2007) In vitro effects of adrenomedullin and calcitonin gene related peptide on the release of serotonin and amino acids from rat dorsal spinal cord. Neurosci Lett 420(3):193–197
Löken LS, Braz JM, Etlin A, Sadeghi M, Bernstein M, Jewell M et al (2021) Contribution of dorsal Horn CGRP-expressing interneurons to mechanical sensitivity. eLife 10:e59751
Ryu P, Gerber G, Murase K, Randic M (1988) Actions of calcitonin gene-related peptide on rat spinal dorsal Horn neurons. Brain Res 441(1–2):357–361
Miletic V, Tan H (1988) Iontophoretic application of calcitonin gene-related peptide produces a slow and prolonged excitation of neurons in the Cat lumbar dorsal Horn. Brain Res 446(1):169–172
Ebersberger A, Charbel Issa P, Vanegas H, Schaible HG (2000) Differential effects of calcitonin gene-related peptide and calcitonin gene-related peptide 8–37 upon responses to N-methyl-d-aspartate or (R,S)-α-amino-3-hydroxy-5-methylisoxazole-4-propionate in spinal nociceptive neurons with knee joint input in the rat. Neuroscience 99(1):171–178
Welch SP, Cooper C, Dewey WL (1988) An investigation of the antinociceptive activity of calcitonin gene-related peptide alone and in combination with morphine: correlation to 45Ca + + uptake by synaptosomes. J Pharmacol Exp Ther 244(1):28–33
Kangrga I, Randic M (1990) Tachykinins and calcitonin gene-related peptide enhance release of endogenous glutamate and aspartate from the rat spinal dorsal Horn slice. J Neurosci 10(6):2026–2038
Oku R, Satoh M, Fujii N, Otaka A, Yajima H, Takagi H (1987) Calcitonin gene-related peptide promotes mechanical nociception by potentiating release of substance P from the spinal dorsal Horn in rats. Brain Res 403(2):350–354
Wei S-Q, Tao Z-Y, Xue Y, Cao D-Y (2020) Peripheral sensitization. Peripheral nerve disorders and treatment.:131– 56
Burstein R, Jakubowski M, Rauch SD (2011) The science of migraine. J Vestib Res 21(6):305–314
Chaban VV (2010) Peripheral sensitization of sensory neurons. Ethn Dis 20(1 Suppl 1):S1
Kung L-H, Gong K, Adedoyin M, Ng J, Bhargava A, Ohara PT et al (2013) Evidence for glutamate as a neuroglial transmitter within sensory ganglia. PLoS ONE 8(7):e68312
Inquimbert P, Bartels K, Babaniyi OB, Barrett LB, Tegeder I, Scholz J (2012) Peripheral nerve injury produces a sustained shift in the balance between glutamate release and uptake in the dorsal Horn of the spinal cord. PAIN® 153(12):2422–2431
Cho YS, Mah W, Youn DH, Kim YS, Ko H-G, Bae JY et al (2023) Increase of glutamate in satellite glial cells of the trigeminal ganglion in a rat model of craniofacial neuropathic pain. Front Neuroanat 17:1302373
Capuano A, De Corato A, Lisi L, Tringali G, Navarra P, Russo CD (2009) Proinflammatory-activated trigeminal satellite cells promote neuronal sensitization: relevance for migraine pathology. Mol Pain 5:1744–8069
Benbow T, Cairns BE (2021) Dysregulation of the peripheral glutamatergic system: A key player in migraine pathogenesis? Cephalalgia 41(11–12):1249–1261
Tai M-LS, Yap JF, Goh CB (2018) Dietary trigger factors of migraine and tension-type headache in a South East Asian country. J Pain Res.:1255–1261
Hindiyeh NA, Zhang N, Farrar M, Banerjee P, Lombard L, Aurora SK (2020) The role of diet and nutrition in migraine triggers and treatment: a systematic literature review. Headache: J Head Face Pain 60(7):1300–1316
Schürks M, Buring JE, Kurth T (2011) Migraine features, associated symptoms and triggers: A principal component analysis in the women’s health study. Cephalalgia 31(7):861–869
Nakamura-Craig M, Gill BK (1991) Effect of neurokinin A, substance P and calcitonin gene related peptide in peripheral hyperalgesia in the rat paw. Neurosci Lett 124(1):49–51
Iyengar S, Ossipov MH, Johnson KW (2017) The role of calcitonin gene–related peptide in peripheral and central pain mechanisms including migraine. Pain 158(4):543–559
Weidner C, Klede M, Rukwied R, Lischetzki G, Neisius U, Schmelz M et al (2000) Acute effects of substance P and calcitonin gene-related peptide in human skin–a Microdialysis study. J Invest Dermatology 115(6):1015–1020
Schmelz M, Petersen LJ (2001) Neurogenic inflammation in human and rodent skin. Physiology 16(1):33–37
Pellesi L, Edvinsson L (2025) Revisiting substance P in migraine: a methodological approach inspired by anti-CGRP and anti-PACAP success. J Headache Pain 26(1):22
Spekker E, Tanaka M, Szabó Á, Vécsei L (2021) Neurogenic inflammation: the participant in migraine and recent advancements in translational research. Biomedicines 10(1):76
Ji R-R, Xu Z-Z, Gao Y-J (2014) Emerging targets in neuroinflammation-driven chronic pain. Nat Rev Drug Discovery 13(7):533–548
Su M, Yu S (2018) Chronic migraine: A process of dysmodulation and sensitization. Mol Pain 14:1744806918767697
Dodick D, Silberstein S (2006) Central sensitization theory of migraine: Clinical implications. Headache.;46(SUPPL. 4):S182-S91
Andreou AP (2022) Glutamate in migraine neurobiology and treatment. In: Pavlovic ZM (ed) Glutamate and neuropsychiatric disorders: current and emerging treatments. Springer International Publishing, Cham, pp 195–228
Selby G, Lance JW, OBSERVATIONS, ON 500 CASES OF MIGRAINE AND ALLIED VASCULAR HEADACHE (1960) J Neurol Neurosurg Psychiatry 23(1):23–32
Burstein R, Cutrer MF, Yarnitsky D (2000) The development of cutaneous allodynia during a migraine attack. Clinical evidence for the sequential recruitment of spinal and supraspinal nociceptive neurons in migraine. Brain 123(8):1703–1709
Landy S, Rice K, Lobo B (2004) Central sensitisation and cutaneous allodynia in migraine: implications for treatment. CNS Drugs 18:337–342
Woolf CJ, Thompson SW (1991) The induction and maintenance of central sensitization is dependent onN-methyl-d-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity States. Pain 44(3):293–299
Jackson DL, Graff CB, Richardson JD, Hargreaves KM (1995) Glutamate participates in the peripheral modulation of thermal hyperalgesia in rats. Eur J Pharmacol 284(3):321–325
Zhou S, Bonasera L, Carlton SM (1996) Peripheral administration of NMDA, AMPA or KA results in pain behaviors in rats. NeuroReport 7(4):895–900
Bereiter DA, Bereiter DF, Hathaway CB (1996) The NMDA receptor antagonist MK-801 reduces Fos-like immunoreactivity in central trigeminal neurons and blocks select endocrine and autonomic responses to corneal stimulation in the rat. Pain 64(1):179–189
Birder LA, Roppolo JR, Erickson VL, de Groat WC (1999) Increased c-fos expression in spinal lumbosacral projection neurons and preganglionic neurons after irritation of the lower urinary tract in the rat. Brain Res 834(1–2):55–65
Thompson S, Woolf C, Sivilotti L (1993) Small-caliber afferent inputs produce a heterosynaptic facilitation of the synaptic responses evoked by primary afferent A-fibers in the neonatal rat spinal cord in vitro. J Neurophysiol 69(6):2116–2128
Neugebauer V, Lücke T, Schaible HG (1994) Requirement of metabotropic glutamate receptors for the generation of inflammation-evoked hyperexcitability in rat spinal cord neurons. Eur J Neurosci 6(7):1179–1186
Young MR, Fleetwood-Walker SM, Dickinson T, Blackburn-Munro G, Sparrow H, Birch PJ et al (1997) Behavioural and electrophysiological evidence supporting a role for group I metabotropic glutamate receptors in the mediation of nociceptive inputs to the rat spinal cord. Brain Res 777(1–2):161–169
Azkue JJ, Liu X-G, Zimmermann M, Sandkühler J (2003) Induction of long-term potentiation of C fibre-evoked spinal field potentials requires recruitment of group I, but not group II/III metabotropic glutamate receptors. Pain 106(3):373–379
Soliman AC, Jonathan S, Coderre TJ (2005) mGlu and NMDA receptor contributions to capsaicin-induced thermal and mechanical hypersensitivity. Neuropharmacology 48(3):325–332
Young M, Fleetwood-Walker S, Mitchell R, Munro F (1994) Evidence for a role of metabotropic glutamate receptors in sustained nociceptive inputs to rat dorsal Horn neurons. Neuropharmacology 33(1):141–144
Zhou X, Liang J, Wang J, Fei Z, Qin G, Zhang D et al (2020) Up-regulation of astrocyte excitatory amino acid transporter 2 alleviates central sensitization in a rat model of chronic migraine. J Neurochem 155(4):370–389
Schieber M, Chandel NS (2014) ROS function in redox signaling and oxidative stress. Curr Biol 24(10):R453–R62
Murugan M, Ling E-A, Kaur C (2013) Glutamate receptors in microglia. CNS & neurological Disorders-Drug targets (Formerly current drug targets-CNS & neurological Disorders). 12(6):773–784
Holton KF (2021) Micronutrients May be a unique weapon against the neurotoxic triad of excitotoxicity, oxidative stress and neuroinflammation: a perspective. Front NeuroSci 15:726457
Zhang X, Zhang W, Wang Y, Zhang Y, Zhang D, Qin G et al (2024) SIRT1-regulated ROS generation activates NMDAR2B phosphorylation to promote central sensitization and allodynia in a male chronic migraine rat model. Front Mol Neurosci 17:1387481
Gao X, Kim HK, Mo Chung J, Chung K (2007) Reactive oxygen species (ROS) are involved in enhancement of NMDA-receptor phosphorylation in animal models of pain. Pain 131(3):262–271
Brenner GJ, Ji RR, Shaffer S, Woolf CJ (2004) Peripheral noxious stimulation induces phosphorylation of the NMDA receptor NR1 subunit at the PKC-dependent site, serine‐896, in spinal cord dorsal Horn neurons. Eur J Neurosci 20(2):375–384
Jones TL, Sorkin LS (2005) Activated PKA and PKC, but not CaMKIIα, are required for AMPA/kainate-mediated pain behavior in the thermal stimulus model. Pain 117(3):259–270
Vergne-Salle P, Bertin P (2021) Chronic pain and neuroinflammation. Joint Bone Spine 88(6):105222
Sun S, Fan Z, Liu X, Wang L, Ge Z (2024) Microglia TREM1-mediated neuroinflammation contributes to central sensitization via the NF-κB pathway in a chronic migraine model. J Headache Pain 25(1):3
Kawasaki Y, Zhang L, Cheng J-K, Ji R-R (2008) Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1β, interleukin-6, and tumor necrosis factor-α in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci 28(20):5189–5194
Zhang L, Berta T, Xu Z-Z, Liu T, Park JY, Ji R-R (2011) TNF-alpha contributes to spinal cord synaptic plasticity and inflammatory pain: distinct role of TNF receptor subtypes 1 and 2. Pain 152(2):419–427
Marquez de Prado B, Hammond DL, Russo AF (2009) Genetic enhancement of calcitonin Gene-Related Peptide-Induced central sensitization to mechanical stimuli in mice. J Pain 10(9):992–1000
Giniatullin R, Nistri A, Fabbretti E (2008) Molecular mechanisms of sensitization of pain-transducing P2X 3 receptors by the migraine mediators CGRP and NGF. Mol Neurobiol 37:83–90
Fabbretti E, D’Arco M, Fabbro A, Simonetti M, Nistri A, Giniatullin R (2006) Delayed upregulation of ATP P2X3 receptors of trigeminal sensory neurons by calcitonin gene-related peptide. J Neurosci 26(23):6163–6171
Khodorova A, Richter J, Vasko MR, Strichartz G (2009) Early and late contributions of glutamate and CGRP to mechanical sensitization by endothelin-1. J Pain 10(7):740–749
Balonov K, Khodorova A, Strichartz GR (2006) Tactile allodynia initiated by local subcutaneous Endothelin-1 is prolonged by activation of TRPV-1 receptors. Experimental Biology Med 231(6):1165–1170
da Cunha JM, Rae GA, Ferreira SH, Cunha FQ (2004) Endothelins induce ETB receptor-mediated mechanical hypernociception in rat hindpaw: roles of cAMP and protein kinase C. Eur J Pharmacol 501(1–3):87–94
Hougaard A, Younis S, Iljazi A, Sugimoto K, Ayata C, Ashina M (2020) Intravenous Endothelin-1 infusion does not induce aura or headache in migraine patients with aura. Headache: J Head Face Pain 60(4):724–734
Chah N, Jones M, Milord S, Al-Eryani K, Enciso R (2021) Efficacy of ketamine in the treatment of migraines and other unspecified primary headache disorders compared to placebo and other interventions: a systematic review. J Dent Anesth Pain Med 21(5):413–429
Li G, Qu B, Zheng T, Duan S, Liu L, Liu ZJ (2024) Role of memantine in adult migraine: A systematic review and network meta-analysis to Ompare memantine with existing migraine preventive treatment medications. Front Pharmacol 15:1496621
Muir KW (2006) Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists. Curr Opin Pharmacol 6(1):53–60
Chan K, MaassenVanDenBrink A (2014) Glutamate receptor antagonists in the management of migraine. Drugs 74(11):1165–1176
Charles A, Flippen C, Romero Reyes M, Brennan KC (2007) Memantine for prevention of migraine: a retrospective study of 60 cases. J Headache Pain 8:248–250
Bigal M, Rapoport A, Sheftell F, Tepper D, Tepper S (2008) Memantine in the preventive treatment of refractory migraine. Headache: J Head Face Pain 48(9):1337–1342
Kaube H, Herzog J, Kaufer T, Dichgans M, Diener H (2000) Aura in some patients with Familial hemiplegic migraine can be stopped by intranasal ketamine. Neurology 55(1):139–141
Afridi SK, Giffin NJ, Kaube H, Goadsby PJ (2013) A randomized controlled trial of intranasal ketamine in migraine with prolonged aura. Neurology 80(7):642–647
Sheth MK, Brand A, Halterman J (2018) Ketamine-induced changes in blood pressure and heart rate in pre-hospital intubated patients. Adv J Graduate Res 3(1):20–33
Morgan CJ, Curran HV (2006) Acute and chronic effects of ketamine upon human memory: a review. Psychopharmacology 188:408–424
Morgan CJA, Curran HV, Drugs tISCo (2012) Ketamine use: a review. Addiction 107(1):27–38
Tennant DR (2018) Review of glutamate intake from both food additive and non-additive sources in the European union. Annals Nutr Metabolism 73(Suppl 5):21–28
Martami F, Holton KF (2023) Targeting glutamate neurotoxicity through dietary manipulation: potential treatment for migraine. Nutrients 15(18):3952
Raffaelli B, Neeb L, Reuter U (2019) Monoclonal antibodies for the prevention of migraine. Expert Opin Biol Ther 19(12):1307–1317
Dodick DW, Lipton RB, Silberstein S, Goadsby PJ, Biondi D, Hirman J et al (2019) Eptinezumab for prevention of chronic migraine: a randomized phase 2b clinical trial. Cephalalgia 39(9):1075–1085
Silberstein SD, Dodick DW, Bigal ME, Yeung PP, Goadsby PJ, Blankenbiller T et al (2017) Fremanezumab for the preventive treatment of chronic migraine. N Engl J Med 377(22):2113–2122
McAllister P, Lamerato L, Krasenbaum LJ, Cohen JM, Tangirala K, Thompson S et al (2021) Real-world impact of fremanezumab on migraine symptoms and resource utilization in the united States. J Headache Pain 22:1–8
Kudrow D, Cady RK, Allan B, Pederson SM, Hirman J, Mehta LR et al (2021) Long-term safety and tolerability of eptinezumab in patients with chronic migraine: a 2-year, open-label, phase 3 trial. BMC Neurol 21:1–12
Xu D, Chen D, Zhu L-n, Tan G, Wang H-j, Zhang Y et al (2019) Safety and tolerability of calcitonin-gene-related peptide binding monoclonal antibodies for the prevention of episodic migraine–a meta-analysis of randomized controlled trials. Cephalalgia 39(9):1164–1179
Torres-Ferrús M, Gallardo VJ, Alpuente A, Caronna E, Gine-Cipres E, Pozo-Rosich P (2021) The impact of anti-CGRP monoclonal antibodies in resistant migraine patients: a real-world evidence observational study. J Neurol.:1–10
Scheffler A, Schenk H, Wurthmann S, Nsaka M, Kleinschnitz C, Glas M et al (2021) CGRP antibody therapy in patients with drug resistant migraine and chronic daily headache: a real-world experience. J Headache Pain 22:1–6
Melo-Carrillo A, Strassman AM, Nir R-R, Schain AJ, Noseda R, Stratton J et al (2017) Fremanezumab—a humanized monoclonal anti-CGRP antibody—inhibits thinly myelinated (Aδ) but not unmyelinated (C) meningeal nociceptors. J Neurosci 37(44):10587–10596
Burstein R, Yamamura H, Malick A, Strassman AM (1998) Chemical stimulation of the intracranial dura induces enhanced responses to facial stimulation in brain stem trigeminal neurons. J Neurophysiol 79(2):964–982
Puledda F, de Boer I, Messina R, Garcia-Azorin D, Portes Souza MN, Al-Karagholi MA-M et al (2024) Worldwide availability of medications for migraine and tension-type headache: A survey of the international headache society. Cephalalgia 44(11):03331024241297688
Sacco S, Bendtsen L, Ashina M, Reuter U, Terwindt G, Mitsikostas D-D et al (2019) European headache federation guideline on the use of monoclonal antibodies acting on the calcitonin gene related peptide or its receptor for migraine prevention. J Headache Pain 20:1–33
Sacco S, Amin FM, Ashina M, Bendtsen L, Deligianni CI, Gil-Gouveia R et al (2022) European headache federation guideline on the use of monoclonal antibodies targeting the calcitonin gene related peptide pathway for migraine prevention–2022 update. J Headache Pain 23(1):67
Uzun S, Frejvall U, Petersson P, Sahin G (2024) Constipation as a possible predictor of poor treatment response in chronic migraine: A retrospective study of anti-calcitonin gene-related peptide (anti-CGRP) monoclonal antibodies and the impact of switching. Cephalalgia Rep 7:25158163241292307
Saely S, Croteau D, Jawidzik L, Brinker A, Kortepeter C (2021) Hypertension: a new safety risk for patients treated with erenumab. Headache: J Head Face Pain 61(1):202–208
Ruiz M, Cocores A, Tosti A, Goadsby PJ, Monteith TS (2023) Alopecia as an emerging adverse event to CGRP monoclonal antibodies: cases series, evaluation of FAERS, and literature review. Cephalalgia 43(2):03331024221143538
Wang Q, Liu J, Sun H, Dong Y, Tan W, Tang Z et al (2024) Adverse event profile of CGRP monoclonal antibodies: findings from the FDA adverse event reporting database. Exp Opin Drug Saf 23(1):107–117
Van Der Arend BWH, Van Veelen N, De Ruijter JET, Olsen MH, MaassenVanDenBrink A, Terwindt GM (2024) Safety considerations in the treatment with anti-CGRP(R) monoclonal antibodies in patients with migraine. Front Neurol.;15
Andress-Rothrock D, King W, Rothrock J (2010) An analysis of migraine triggers in a clinic‐based population. Headache: J Head Face Pain 50(8):1366–1370
Fukui PT, Gonçalves TRT, Strabelli CG, Lucchino NMF, Matos FC, Santos JPM et al (2008) Trigger factors in migraine patients. Arq Neuropsiquiatr 66:494–499
Marmura MJ (2018) Triggers, protectors, and predictors in episodic migraine. Curr Pain Headache Rep 22:1–9
Schulte LH, Jürgens TP, May A (2015) Photo-, osmo-and phonophobia in the premonitory phase of migraine: mistaking symptoms for triggers? J Headache Pain 16:1–5
Cottier KE, Galloway EA, Calabrese EC, Tome ME, Liktor-Busa E, Kim J et al (2018) Loss of blood-brain barrier integrity in a KCl-induced model of episodic headache enhances CNS drug delivery. Eneuro.;5(4)
Morselli P, Garattini S (1970) Monosodium glutamate and the Chinese restaurant syndrome. Nature 227(5258):611–612
Schaumburg HH, Byck R, Gerstl R, Mashman JH (1969) Monosodium L-glutamate: its Pharmacology and role in the Chinese restaurant syndrome. Science 163(3869):826–828
Scopp AL (1991) MSG and hydrolyzed vegetable protein induced headache: review and case studies. Headache: J Head Face Pain 31(2):107–110
Shimada A, Cairns BE, Vad N, Ulriksen K, Pedersen AML, Svensson P et al (2013) Headache and mechanical sensitization of human pericranial muscles after repeated intake of monosodium glutamate (MSG). J Headache Pain 14:1–9
Holton KF, Taren DL, Bennett RM, Jones KD (2010) Novel dietary intervention produces significant improvements in fibromyalgia patients with irritable bowel syndrome. FASEB J 24(S1):56423–56423
Cairns BE, Dong X, Mann MK, Svensson P, Sessle BJ, Arendt-Nielsen L et al (2007) Systemic administration of monosodium glutamate elevates intramuscular glutamate levels and sensitizes rat masseter muscle afferent fibers. Pain 132(1):33–41
Blachier F, Guihot-Joubrel G, Vaugelade P, Le Boucher J, Bernard F, Duée P-H et al (1999) Portal hyperglutamatemia after dietary supplementation with monosodium glutamate in pigs. Digestion 60(4):349–357
Additives EP, oF F, NSat, Mortensen A, Aguilar F, Crebelli R, Di Domenico A et al (2017) Re-evaluation of glutamic acid (E 620), sodium glutamate (E 621), potassium glutamate (E 622), calcium glutamate (E 623), ammonium glutamate (E 624) and magnesium glutamate (E 625) as food additives. EFSA J 15(7):e04910
Gazerani P (2021) A bidirectional view of migraine and diet relationship. Neuropsychiatr Dis Treat.:435–451
Hong JB, Lange KS, Overeem LH, Triller P, Raffaelli B, Reuter U (2023) A scoping review and Meta-Analysis of Anti-CGRP monoclonal antibodies: predicting response. Pharmaceuticals 16(7):934
Hosseini-Zare MS, Gu F, Abdulla A, Powell S, Žiburkus J (2017) Effects of experimental traumatic brain injury and impaired glutamate transport on cortical spreading depression. Exp Neurol 295:155–161
Eikermann-Haerter K, Ayata C (2010) Cortical spreading depression and migraine. Curr Neurol Neurosci Rep 10:167–173
Acknowledgements
Not applicable.
Funding
The authors did not receive funding from any organization for the submitted work.
Author information
Authors and Affiliations
Contributions
FM conducted the literature review and drafted the manuscript. KH provided guidance on the topic, critically edited and revised the manuscript. Both authors approved the final version.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Martami, F., Holton, K.F. Unmasking the relationship between CGRP and glutamate: from peripheral excitation to central sensitization in migraine. J Headache Pain 26, 101 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10194-025-02043-x
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10194-025-02043-x