Skip to main content
Erschienen in: The Journal of Headache and Pain 1/2017

Open Access 01.12.2017 | Review article

The KATP channel in migraine pathophysiology: a novel therapeutic target for migraine

verfasst von: Mohammad Al-Mahdi Al-Karagholi, Jakob Møller Hansen, Johanne Severinsen, Inger Jansen-Olesen, Messoud Ashina

Erschienen in: The Journal of Headache and Pain | Ausgabe 1/2017

Abstract

Background

To review the distribution and function of KATP channels, describe the use of KATP channels openers in clinical trials and make the case that these channels may play a role in headache and migraine.

Discussion

KATP channels are widely present in the trigeminovascular system and play an important role in the regulation of tone in cerebral and meningeal arteries. Clinical trials using synthetic KATP channel openers report headache as a prevalent-side effect in non-migraine sufferers, indicating that KATP channel opening may cause headache, possibly due to vascular mechanisms. Whether KATP channel openers can provoke migraine in migraine sufferers is not known.

Conclusion

We suggest that KATP channels may play an important role in migraine pathogenesis and could be a potential novel therapeutic anti-migraine target.
Abkürzungen
ABC transporter
ATP-binding cassette transporter
BA
Basilar artery
CGRP
Calcitonin gene-related peptide
CSD
Cortical spread depression
DRG
Dorsal root ganglia
KATP channel
Adenosine 5′-triphosphate-sensitive K+ channel
Kir
K+ inwardly rectifying
MCA
Middle cerebral artery
Mg-ADP
Magnesium adenosine diphosphate
MMA
Middle meningeal artery
NBD
Nucleotide binding domains
NO
Nitric oxide
PACAP
Pituitary adenylate cyclase activating polypeptide
PGI2
Prostacyclin
SUR
Sulfonylurea receptor
TG
Trigeminal ganglion,
TNC
Trigeminal nucleus caudatus
VOCC
Voltage-operated Ca2+-channels

Introduction

Adenosine 5′-triphosphate-sensitive K+ (KATP) channel openers have been used in clinical trials for the treatment of hypertension and asthma. The most common side effect mentioned during treatment with KATP channel openers was headache (62, 64, 66–79) (Tables 2 and 3). However, only little attention has been focused on the role of KATP channels in migraine pathophysiology.
KATP channels were originally identified in cardiomyocytes [1], but have also been found in several tissues, including pancreatic α- and ß-cells, smooth muscle, skeletal muscle and central neurons [2, 3]. The channels belong to the family of inwardly rectifying K+ channels that are inhibited at physiological intracellular levels ATP/ADP ratio. When intracellular ATP is reduced under conditions of metabolic challenges they open. KATP channels are critical in regulating insulin secretion, controlling vascular tone, and protecting cells against metabolic stress [2, 4, 5].
Over the past three decades, some preclinical evidence has emerged indicating that KATP channels may play an important role in migraine pathophysiology. In particular, the vasodilation effect of KATP channels is relevant, since it is has been established that endogenous neurotransmitters that trigger migraine attacks are often associated with dilation of cranial arteries [6].
Here we review preclinical and clinical studies on KATP channels and discuss the KATP channel as a novel therapeutic target for migraine treatment.

Molecular structure and isoforms

The KATP channel is a hetero-octameric complex that consists of four pore-forming K+ inwardly rectifying (Kir) subunits and four regulatory sulfonylurea receptor (SUR) subunits [7].
The Kir6.x subunit exists in two isoforms, Kir6.1 and Kir6.2. The SUR subunit belongs to the ATP-binding cassette (ABC) transporter family, regulated by sulfonylurea, with three isoforms, SUR1, SUR2A, and SUR2B [7, 8].
KATPchannels have specific tissue expression with different compositions of Kir6.x and SUR subunits which lead to distinct functional properties (Figs. 1 and 2 and Table 1).
Table 1
Distribution of KATP channels
Subtypes of KATPchannels
Tissue expression
Migraine related structures
Kir6.2/SUR1
Pancreas and brain
DRG, TG and TNC from rats (20–24, 26).
Kir6.2/SUR2A
Cardiac and skeletal muscle
 
Kir6.2/SUR2B
Smooth muscle
DRG, TG, TNC, BA and MCA from rats(20–24, 26).
Kir6.1/SUR2B
Smooth muscle
MMA from rats, pigs and human; MCA from rats and pigs; BA, DRG, TG and TNC from rats (20–24, 26).
DRG Dorsal root ganglia, TG trigeminal ganglion, TNC trigeminal nucleus caudatus, BA basilar artery, MMA middle meningeal artery, MCA middle cerebral artery

Channel function

KATP channel activity is controlled by changes in concentrations of intracellular ATP and magnesium adenosine diphosphate (Mg-ADP). KATP channels couple the metabolic state of the cell to the membrane potential and thus play a crucial role in many tissues under both physiological and pathological conditions [9]. K+ channels participate in the regulation of vascular tone, including cerebral arteries [10]. When intracellular ATP is reduced, KATP channels become activated; K+ efflux hyperpolarize the membrane and close voltage-operated Ca2+-channels (VOCC). The result is a decrease in cytosolic Ca2+ concentration followed by relaxation of vascular smooth muscle cells and an increase in blood flow [11]. The same applies if cells are exposed to metabolic stress such as ischemia or hypoglycemia [12]. Closure of K+ channels leads to membrane depolarization and constriction of the vessels [11]. In addition an increase in intracellular cAMP and cGMP levels activate KATP channels to produce vasodilation [11]. Synthetic KATP channel openers (like levcromakalim and cromakalim) and blockers (like glibenclamide, second generation of sulfonylurea and PNU37883A) directly activate or inhibit the vascular KATP channels, respectively [9] (Fig. 3).

Distribution of KATP channels in migraine related structures

Intracranial arteries

KATP channels are present and functional in intracranial arteries [1315]. They are found in vascular smooth muscle cells and vascular endothelial cells [16, 17]. In rat cerebral arteries, the distribution of KATP channels varies with vessel size and brain region [18]. Real time polymerase chain reaction (RT-PCR) analysis revealed Kir6.1 and SUR2B subunits in middle meningeal artery (MMA) and middle cerebral artery (MCA) in rats and pigs [19, 20]. This profile of KATP channels is also identified in human MMA [21] (Table 1).

Trigeminal ganglion and trigeminal nucleus caudalis

Kir6.1, Kir6.2, SUR1 and SUR2 are expressed in the trigeminal ganglion and trigeminal nucleus caudalis [22] (Table 1). In trigeminal neurons Kir 6.1 and Kir 6.2 immunoreactivity were expressed in cells with all soma sizes in all three divisions of the trigeminal ganglion [23].

KATP channels openers and migraine signaling pathways

A number of endogenous vasoactive signaling molecules have been implicated in migraine [6], and KATP channels may interact with these molecules.

Nitric oxide (NO)

In humans, infusion of the NO donor, glyceryl trinitrate, and inhibition of the breakdown of cGMP by sildenafil [24] provoke migraine attacks in migraineurs [2527]. The NO-cGMP signaling pathway is involved in the relaxation of vascular smooth muscle [28]. In vitro studies with cerebral arteries isolated from rat and piglet and extra-cerebral arteries from rabbit reported that activation (opening) of KATP channels contributed to both cAMP- and cGMP-mediated vasodilation [2931]. Yuan et al. [32] reported that sildenafil-induced vasodilation in porcine retinal arterioles was significantly inhibited by glibenclamide and suggested that cGMP signaling triggers opening of KATP channels. In contrast, NO-induced dural and pial artery dilation in rats was not attenuated by the KATP channel blocker, glibenclamide [33]. Together, these data suggest that interspecies differences are likely to explain the discrepancy in findings of the role of KATP channels in NO-induced vasodilation.
CGRP is one of the most potent endogenous vasodilators and major arteries in the intracranial circulation of man and animals are innervated by CGRP-containing nerve fibers [3436]. Efficacy of CGRP antagonism is established in acute [37, 38] and preventive treatment of migraine [39]. CGRP activates vascular smooth muscle KATP channels indirectly through adenylate cyclase and protein kinase A (PKA) phosphorylation (Fig. 4) [4043]. In rats, CGRP-induced dilation of the dural and pial arteries in vivo was shown to be inhibited by glibenclamide [33], but KATP channel openers do not interact with CGRP release in trigeminal ganglion and trigeminal nucleus caudalis [22]. This suggests that KATP channels are involved in CGRP-induced intracranial vasodilation.

Pituitary adenylate cyclase activating polypeptide (PACAP)

Pituitary adenylate cyclase activating polypeptide (PACAP) is a potent endothelium independent vasodilator of various vascular beds, including cerebral arteries [44, 45]. In vivo and in vitro studies have demonstrated that PACAP dilates cranial arteries in different species, e.g. human cerebral arteries [34, 46, 47], pig pia artery, canine basilar artery, cat cerebral arteries, rabbit posterior cerebral arteries and rat middle cerebral arteries [4852]. Emerging data suggest that PACAP or it receptors are a promising target for migraine therapeutics [53]. PACAP has three types of receptors; Pituitary adenylate cyclase PAC1 (pituitary adenylate cyclase receptor 1), VPAC1 (vasoactive intestinal peptide and pituitary adenylate cyclase receptor 1) and VPAC2 (vasoactive intestinal peptide and pituitary adenylate cyclase receptor 2) [54] the two latter ones are also activated by vasoactive intestinal peptide and all three receptors are found in cerebral artery smooth muscle cells [55]. Through these receptors, PACAP leads to an increase in intracellular cAMP, which activates PKA and produces vasodilation by several mechanisms including activation of KATP channels (Fig. 4) [45]. Interestingly, glibenclamide could partially inhibit PACAP induced vasodilation in cerebral, coronary and pulmonary arteries, suggesting that PACAP may also activate KATP channels [44, 45].

Prostaglandins

Prostacyclin (PGI2) activates and sensitizes meningeal sensory afferents, and provokes immediate migraine-like attacks in migraine sufferers [56]. PGI2 also increases KATP channel activity in vascular smooth muscle preparations by cAMP-dependent PKA activation [57] (Fig. 4).

Headache induced by KATP channels openers

In the late 80’s there was a tremendous interest in developing novel KATP channel openers for hypertension, angina pectoris and asthma. Three pharmacological drugs were developed, pinacidil, nicorandil and levcromakalim. One of most common adverse events after treatment reported in these studies was headache [5863].
Six clinical trials with pinacidil have been published for treatment of essential hypertension. Between 7% and 21% of the patients reported headache as an adverse effect (Table 2).
Table 2
Headache incidences registered during randomized controlled trials (RCT) and open label clinical trials with pinacidil
Paper
Study design
Dose (daily)
Indication
No. of patients
Headache No.
Muiesan et al. 1985, Eur. J. Clin. Pharmacol [86].
RCT
30–75 mg
Essential hypertension
30
2 (7%)
Laher & Hickey 1985, J. Int. Med. Res [87].
Open label
12.5 mg
Healthy volunteers
12
1 (8%)
D’Arcy et al. 1985, Eur. J. Clin. Pharmacol [88].
Open label
20–100 mg
Essential hypertension
23
4 (17%)
Zachariah et al. 1986, Eur. J. Clin. Pharmacol [89].
RCT
62 mg (mean)
Essential hypertension
23
----
Sterndorff & Johansen 1988, Acta Med. Scand [90].
RCT
25–100 mg
Essential hypertension
71
7 (10%)
Goldberg 1988, J. Cardiovasc. Pharmacol [91].
RCT
25–100 mg
Essential hypertension
145
31 (21%)
Nicorandil was tested for the treatment of angina pectoris and ischemic heart disease. 23% to 88% of the patients reported headache as an adverse event (Table 3). The high incidence of headache is likely due to the mixed KATP channel opener and NO donor properties of nicorandil which thus cause vasodilation via two separate mechanisms.
Table 3
Headache incidences registered during randomized controlled trials (RCT) and open label clinical trials with nicorandil
Paper
Study design
Dose (daily)
Indication
No. of patients
Headache No.
Camm & Maltz, 1989, Am. J. Cardiol [92].
RCT
20–60 mg
Angina pectoris
8
20 mg 50%
40 mg 88%
60 mg 67%
Raftery et al. 1993, Eur. Heart Journal [93].
RCT
20 mg and 40 mg
Angina pectoris
18
11 (61%)
Roland 1993, Eur. Heart Journal [94].
Review
10–80 mg
Angina pectoris
1680
36%
Wolf et al. 1993, Eur.J.Clin.Pharmacol [95].
RCT
20–200 μg i.v.
Healthy volunteers
48
19 (40%)
Witchitz & Darmaon, 1995, Cardiovasc. Drugs& Therap [96].
Open label
20–40 mg
Angina pectoris
197
45 (23%)
Dunn et al. 1999, Pharmacoepidemiology and Drug safety [97].
Prescription-event monitoring (PEM) study
Varying
Angina pectoris & ischemic heart disease
13,260
477 (4%)
Levcromakalim was investigated for the treatment of asthma and essential hypertension. In these studies between 29% and 76% of the patients reported headache as an adverse event (Table 4).
Table 4
Headache incidences registered during randomized controlled trials (RCT) and open label clinical trials with levcromakalim
Paper
Study design
Dose (daily)
Indication
No. of patients
Headache No.
Singer et al. 1989, J. Hypertens [98].
RCT
1.5 mg
Essential hypertension
8
4 (50%)
Williams et al. 1990, Lancet [60].
RCT
1.5 mg
Asthma
16
10 (62%)
Kidney et al. 1993, Thorax [62].
RCT
0.125–0.5 mg
Asthma
25
19 (76%)
Suzuki et al. 1995, Arzneim.-Forsch./Drug Res [99].
Open label
0.5–1.0 mg
Essential hypertension
14
4 (29%)
The selective synthetic KATP channel openers levcromakalim and pinacidil have been shown to induce dilation in rat cranial arteries [13, 15, 19] and in isolated human cerebral arteries [64]. Moreover, the arterial dilation can be inhibited by synthetic KATP channel blockers like glibenclamide [10, 33] and PNU37883A [21, 65] (Fig. 3). These findings suggest that high incidences of headache could be due to vasoactive effect of the KATP channel openers in pain-sensitive extra- and/or intracerebral arteries.

Discussion and future perspectives

KATPchannels are expressed in migraine-related structures such as the cranial arteries, TG and TNC [1822, 66]. KATP channels are also connected to a number of key molecules in migraine pathogenesis, particularly nitric oxide, CGRP, PACAP and PGI2 known to provoke migraine attacks [56, 6771]. Therefore, the KATP channels are interesting in migraine context.
Human experimental models have demonstrated that the activation of the cAMP and cGMP pathways can trigger headache in healthy volunteers and migraine attacks in migraine sufferers [6, 71, 72]. The cAMP and cGMP signaling pathways are crucial in the activation of KATP channels, which result in the relaxation of smooth muscle [2931]. Furthermore, synthetic KATP channel openers like levcromakalim and pinacidil trigger headache in non-migraine patients [5863]. Although a detailed description of levcromakalim- and pinacidil-induced headache and accompanying symptoms are lacking, these data support a role of KATP channels in migraine headache. Because KATP channel openers were tested for other indications, there are no available data on the potential migraine-inducing effects of pinacidil and levcromakalim in migraine patients. It is conceivable that both headache and migraine are underreported as adverse events, as was found for the phosphodiesterase inhibitors, cilostazol and sildenafil [73, 74].
In addition to the vasoactive effects, the KATP channels might also tap into other parts of the migraine cascade. For a number of patients, migraine attacks are associated with transient focal neurological symptoms called the aura [75], possibly caused by cortical spread depression (CSD) [76]. During CSD K+ conductance is increased, and CSD may be inhibited by Kir antagonist [77]. The fact that KATP channels open under cellular stress, as seen during long lasting depolarizations, could provide a link between KATP channels, CSD and migraine aura.
With regard to the migraine pain, it is worth noting that KATP channels are also found in peripheral nociceptive fibers [78] and activation of these channels play a crucial role in anti-nociception at both spinal and supra-spinal levels [23, 79]. The exact role of these findings in the headache induced by KATP channel openers is unknown.
If KATP channel openers are in fact able to trigger migraine, the next step to consider is whether KATP channel antagonists can relieve migraine. KATP blockers for the treatment of migraine should be selective for the Kir6.1/SUR2B subtype because of its dominant presence in vascular tissue (Table 1). The necessity of a subtype specific blocker is unavoidable because of occurrence of different subtypes in different tissues. Glibenclamide cannot be used due to its high affinity to the Kir6.2/SUR1 subtype of KATP channels present in the pancreas with hypoglycemia as a side effect [80]. PNU-37883A is a Kir6.1 selective KATP channel blocker that was originally developed as a diuretic drug [81, 82]. The drug was not approved to human studies because of its cardiac depressant activity in animal studies [83]. This precludes further clinical development of PNU-37883A due to possible serious adverse events but may not exclude further investigations in other blockers against Kir6.1 subunit because it is not clear if all blockers against Kir6.1 subunit have non-favorable effects. These findings indicate that the SUR2B subunit and the Kir6.1 subunit should be a potential target for the treatment of migraine, but proof of concept studies are needed to examine this hypothesis.

Conclusion

Emerging evidence suggests that KATP channels could be involved in the pathophysiology of migraine. KATP channels exist in structures which are believed to be linked to the pathophysiology of migraine, including cerebral and meningeal arteries and the trigeminal system [1922]. It is established that the cAMP signaling pathway and possibly cGMP signaling pathway are involved in the activation of KATP channels [2931]. This is interesting in migraine contexts, as the two signaling pathways are likely to be crucial in the development of a migraine attack.
We suggest that the presented clinical and theoretical evidence support further studies of KATP channel openers in migraine context. Future human studies will help clarify the role of KATP channels in the pathophysiology of migraine.

Funding

This article was supported by the Lundbeck Foundation.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
5.
Zurück zum Zitat Saito T, Fujiwara Y, Fujiwara R, et al (2002) Experimental biology 2001 symposium potassium channels that regulate vascular tone : which are the important players ? ROLE OF AUGMENTED EXPRESSION OF INTERMEDIATE- CONDUCTANCE CA 2 + −ACTIVATED K + CHANNELS IN. 324–329 Saito T, Fujiwara Y, Fujiwara R, et al (2002) Experimental biology 2001 symposium potassium channels that regulate vascular tone : which are the important players ? ROLE OF AUGMENTED EXPRESSION OF INTERMEDIATE- CONDUCTANCE CA 2 + −ACTIVATED K + CHANNELS IN. 324–329
11.
Zurück zum Zitat Chrissobolis S, Sobey CG (2003) Inwardly rectifying potassium channels in the regulation of vascular tone. Curr Drug Targets 4:281–289CrossRefPubMed Chrissobolis S, Sobey CG (2003) Inwardly rectifying potassium channels in the regulation of vascular tone. Curr Drug Targets 4:281–289CrossRefPubMed
14.
Zurück zum Zitat Kitazono T, Faraci FM, Taguchi H, Heistad DD (1995) Role of potassium channels in cerebral blood vessels. Stroke 26:1713–1723CrossRefPubMed Kitazono T, Faraci FM, Taguchi H, Heistad DD (1995) Role of potassium channels in cerebral blood vessels. Stroke 26:1713–1723CrossRefPubMed
15.
16.
Zurück zum Zitat Janigro D, West GA, Gordon EL, Winn HR (1993) ATP-sensitive K+ channels in rat aorta and brain microvascular endothelial cells. Am J Phys. 265:C812–C821 Janigro D, West GA, Gordon EL, Winn HR (1993) ATP-sensitive K+ channels in rat aorta and brain microvascular endothelial cells. Am J Phys. 265:C812–C821
17.
Zurück zum Zitat Faraci FM, Heistad DD (1998) Regulation of the Cerebral Circulation: Role of Endothelium and Potassium Channels. Physiol Rev. 78:53–97 Faraci FM, Heistad DD (1998) Regulation of the Cerebral Circulation: Role of Endothelium and Potassium Channels. Physiol Rev. 78:53–97
27.
Zurück zum Zitat Olesen J, Iversen HK, Thomsen LL (1993) Nitric oxide supersensitivity: a possible molecular mechanism of migraine pain. Neuroreport 4:1027–1030CrossRefPubMed Olesen J, Iversen HK, Thomsen LL (1993) Nitric oxide supersensitivity: a possible molecular mechanism of migraine pain. Neuroreport 4:1027–1030CrossRefPubMed
28.
Zurück zum Zitat Niehaus L, Gottschalk S, Weber U (1998) Effect of drug-induced vasodilatation of basal brain arteries with nitroglycerin on blood flow velocity and volume flow in the middle cerebral artery. Ultraschall Med 19:225–229. doi:10.1055/s-2007-1000495 CrossRefPubMed Niehaus L, Gottschalk S, Weber U (1998) Effect of drug-induced vasodilatation of basal brain arteries with nitroglycerin on blood flow velocity and volume flow in the middle cerebral artery. Ultraschall Med 19:225–229. doi:10.​1055/​s-2007-1000495 CrossRefPubMed
29.
Zurück zum Zitat Armstead WM (1996) Role of ATP-sensitive K+ channels in cGMP-mediated pial artery vasodilation. Am J Phys 270:H423–H426 Armstead WM (1996) Role of ATP-sensitive K+ channels in cGMP-mediated pial artery vasodilation. Am J Phys 270:H423–H426
30.
Zurück zum Zitat Hempelmann RG, Ziegler A, Mehdorn HM (2001) Role of potassium channels in the relaxation induced by the nitric oxide ( NO ) donor DEA / NO in the isolated rat basilar artery. Neurosci Lett 313:21–24CrossRefPubMed Hempelmann RG, Ziegler A, Mehdorn HM (2001) Role of potassium channels in the relaxation induced by the nitric oxide ( NO ) donor DEA / NO in the isolated rat basilar artery. Neurosci Lett 313:21–24CrossRefPubMed
32.
Zurück zum Zitat Yuan Z, Hein TW, Rosa RH, Kuo L (2008) Sildenafil (viagra) evokes retinal arteriolar dilation: dual pathways via NOS activation and phosphodiesterase inhibition. Investig Ophthalmol Vis Sci 49:720–725. doi:10.1167/iovs.07-1208 CrossRef Yuan Z, Hein TW, Rosa RH, Kuo L (2008) Sildenafil (viagra) evokes retinal arteriolar dilation: dual pathways via NOS activation and phosphodiesterase inhibition. Investig Ophthalmol Vis Sci 49:720–725. doi:10.​1167/​iovs.​07-1208 CrossRef
35.
37.
Zurück zum Zitat Olesen J, Diener H, Husstedt IW, et al (2004) Calcitonin gene–related peptide receptor antagonist BIBN 4096 BS for the acute treatment of migraine. Society 1104–1110 Olesen J, Diener H, Husstedt IW, et al (2004) Calcitonin gene–related peptide receptor antagonist BIBN 4096 BS for the acute treatment of migraine. Society 1104–1110
38.
Zurück zum Zitat Ho TW, Ferrari MD, Dodick DW et al (2008) Efficacy and tolerability of MK-0974 (telcagepant), a new oral antagonist of calcitonin gene-related peptide receptor, compared with zolmitriptan for acute migraine: a randomised, placebo-controlled, parallel-treatment trial. Lancet 372:2115–2123. doi:10.1016/S0140-6736(08)61626-8 CrossRefPubMed Ho TW, Ferrari MD, Dodick DW et al (2008) Efficacy and tolerability of MK-0974 (telcagepant), a new oral antagonist of calcitonin gene-related peptide receptor, compared with zolmitriptan for acute migraine: a randomised, placebo-controlled, parallel-treatment trial. Lancet 372:2115–2123. doi:10.​1016/​S0140-6736(08)61626-8 CrossRefPubMed
40.
Zurück zum Zitat Nelson MT, Huang Y, Brayden JE et al (1990) Arterial dilations in response to calcitonin gene-related peptide involve activation of K+ channels. Nature 344:770–773. doi:10.1038/344770a0 CrossRefPubMed Nelson MT, Huang Y, Brayden JE et al (1990) Arterial dilations in response to calcitonin gene-related peptide involve activation of K+ channels. Nature 344:770–773. doi:10.​1038/​344770a0 CrossRefPubMed
42.
Zurück zum Zitat Nakaya Y, City T (1995) Calcitonin gene-related peptide activates the K + channels of vascular smooth muscle cells via adenylate cyclase. 332–336 Nakaya Y, City T (1995) Calcitonin gene-related peptide activates the K + channels of vascular smooth muscle cells via adenylate cyclase. 332–336
43.
Zurück zum Zitat Wellman GC, Quayle JM, Standen NB (1998) ATP-sensitive K+ channel activation by calcitonin gene-related peptide and protein kinase a in pig coronary arterial smooth muscle. J Physiol. 507(1):117–129 Wellman GC, Quayle JM, Standen NB (1998) ATP-sensitive K+ channel activation by calcitonin gene-related peptide and protein kinase a in pig coronary arterial smooth muscle. J Physiol. 507(1):117–129
44.
Zurück zum Zitat Bruch L, Rubel S, Kästner A, et al (1998) Pituitary adenylate cyclase activating peptides relax human pulmonary arteries by opening of K ATP and K Ca channels. 586–587 Bruch L, Rubel S, Kästner A, et al (1998) Pituitary adenylate cyclase activating peptides relax human pulmonary arteries by opening of K ATP and K Ca channels. 586–587
46.
Zurück zum Zitat Amin FM, Asghar MS, Guo S, et al (2011) Headache and prolonged dilatation of the middle meningeal artery by PACAP38 in healthy volunteers. 32:140–149. doi:10.1177/0333102411431333 Amin FM, Asghar MS, Guo S, et al (2011) Headache and prolonged dilatation of the middle meningeal artery by PACAP38 in healthy volunteers. 32:140–149. doi:10.​1177/​0333102411431333​
47.
Zurück zum Zitat Amin FM, Hougaard A, Schytz HW et al (2014) Investigation of the pathophysiological mechanisms of migraine attacks induced by pituitary adenylate cyclase-activating polypeptide-38. Brain 137:779–794. doi:10.1093/brain/awt369 CrossRefPubMed Amin FM, Hougaard A, Schytz HW et al (2014) Investigation of the pathophysiological mechanisms of migraine attacks induced by pituitary adenylate cyclase-activating polypeptide-38. Brain 137:779–794. doi:10.​1093/​brain/​awt369 CrossRefPubMed
50.
Zurück zum Zitat Uddman R, Goadsby PJ, Jansen I, Edvinsson L (1993) PACAP, a VIP-like peptide: immunohistochemical localization and effect upon cat pial arteries and cerebral blood flow. J Cereb Blood Flow Metab 13:291–297. doi:10.1038/jcbfm.1993.36 CrossRefPubMed Uddman R, Goadsby PJ, Jansen I, Edvinsson L (1993) PACAP, a VIP-like peptide: immunohistochemical localization and effect upon cat pial arteries and cerebral blood flow. J Cereb Blood Flow Metab 13:291–297. doi:10.​1038/​jcbfm.​1993.​36 CrossRefPubMed
51.
Zurück zum Zitat Tong S, Parfenova H, Shibata M et al (1993) Pituitary adenylate cyclase-activating polypeptide dilates cerebral arterioles of newborn pigs. Proc Soc Exp Biol Med 203:343–347CrossRefPubMed Tong S, Parfenova H, Shibata M et al (1993) Pituitary adenylate cyclase-activating polypeptide dilates cerebral arterioles of newborn pigs. Proc Soc Exp Biol Med 203:343–347CrossRefPubMed
52.
Zurück zum Zitat Seki Y, Suzuki Y, Baskaya MK et al (1995) The effects of pituitary adenylate cyclase-activating polypeptide on cerebral arteries and vertebral artery blood flow in anesthetized dogs. Eur J Pharmacol 275:259–266CrossRefPubMed Seki Y, Suzuki Y, Baskaya MK et al (1995) The effects of pituitary adenylate cyclase-activating polypeptide on cerebral arteries and vertebral artery blood flow in anesthetized dogs. Eur J Pharmacol 275:259–266CrossRefPubMed
54.
Zurück zum Zitat Vaudry D, Falluel-morel A, Bourgault S et al (2009) Pituitary Adenylate Cyclase-activating polypeptide and its receptors : 20 years after the discovery. Pept Res 61:283–357. doi:10.1124/pr.109.001370.283 Vaudry D, Falluel-morel A, Bourgault S et al (2009) Pituitary Adenylate Cyclase-activating polypeptide and its receptors : 20 years after the discovery. Pept Res 61:283–357. doi:10.​1124/​pr.​109.​001370.​283
58.
Zurück zum Zitat Thomas P, Dixon MS, Winterton SJ, Sheridan DJ (1990) Acute haemodynamic effects of cromakalim in patients with angina pectoris. Br J Clin Pharmacol 29:325–331CrossRefPubMedPubMedCentral Thomas P, Dixon MS, Winterton SJ, Sheridan DJ (1990) Acute haemodynamic effects of cromakalim in patients with angina pectoris. Br J Clin Pharmacol 29:325–331CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Antihypertensive Effect of Levcromakalim in patients with essential hypertension.pdf Antihypertensive Effect of Levcromakalim in patients with essential hypertension.pdf
61.
Zurück zum Zitat von Nguyen P, Holliwell DL, Davis A et al (1991) Effects of the potassium channel activator, cromakalim, on arterial and cardiac responses to norepinephrine, angiotensin II, and isoproterenol in normotensive men. J Cardiovasc Pharmacol 18:797–806CrossRef von Nguyen P, Holliwell DL, Davis A et al (1991) Effects of the potassium channel activator, cromakalim, on arterial and cardiac responses to norepinephrine, angiotensin II, and isoproterenol in normotensive men. J Cardiovasc Pharmacol 18:797–806CrossRef
63.
64.
Zurück zum Zitat Hempelmann RG, Barth HL, Mehdorn HM et al (1995) Effects of potassium channel openers in isolated human cerebral arteries. Neurosurgery 37:1146–1153CrossRefPubMed Hempelmann RG, Barth HL, Mehdorn HM et al (1995) Effects of potassium channel openers in isolated human cerebral arteries. Neurosurgery 37:1146–1153CrossRefPubMed
67.
Zurück zum Zitat Olesen J, Jansen-Olesen I (2000) Nitric oxide mechanisms in migraine. Pathol Biol (Paris) 48:648–657 Olesen J, Jansen-Olesen I (2000) Nitric oxide mechanisms in migraine. Pathol Biol (Paris) 48:648–657
72.
Zurück zum Zitat 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 336:403–405. doi:10.1038/332141a0 CrossRef 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 336:403–405. doi:10.​1038/​332141a0 CrossRef
76.
Zurück zum Zitat Lauritzen M (1994) Pathophysiology of the migraine aura. The spreading depression theory. Brain 117(Pt 1):199–210CrossRefPubMed Lauritzen M (1994) Pathophysiology of the migraine aura. The spreading depression theory. Brain 117(Pt 1):199–210CrossRefPubMed
78.
Zurück zum Zitat Staurengo-Ferrari L, Zarpelon AC, Longhi-Balbinot DT et al (2014) Nitroxyl inhibits overt pain-like behavior in mice: role of cGMP/PKG/ATP-sensitive potassium channel signaling pathway. Pharmacol Reports 66:691–698. doi:10.1016/j.pharep.2014.04.003 CrossRef Staurengo-Ferrari L, Zarpelon AC, Longhi-Balbinot DT et al (2014) Nitroxyl inhibits overt pain-like behavior in mice: role of cGMP/PKG/ATP-sensitive potassium channel signaling pathway. Pharmacol Reports 66:691–698. doi:10.​1016/​j.​pharep.​2014.​04.​003 CrossRef
79.
Zurück zum Zitat North RA, Williams JT, Surprenant A, Christie MJ (1987) Mu and delta receptors belong to a family of receptors that are coupled to potassium channels. Neurobiology 84:5487–5491 North RA, Williams JT, Surprenant A, Christie MJ (1987) Mu and delta receptors belong to a family of receptors that are coupled to potassium channels. Neurobiology 84:5487–5491
81.
Zurück zum Zitat Perricone SC, Humphrey SJ, Skaletzky LL, et al (1994) In rats and dogs. 3693–3700 Perricone SC, Humphrey SJ, Skaletzky LL, et al (1994) In rats and dogs. 3693–3700
82.
Zurück zum Zitat Meisheri KD, Humphrey SJ, Khan SA et al (1993) 4-morpholinecarboximidine-N-1-adamantyl-N’-cyclohexylhydrochloride (U-37883A): pharmacological characterization of a novel antagonist of vascular ATP-sensitive K+ channel openers. J Pharmacol Exp Ther 266:655–665PubMed Meisheri KD, Humphrey SJ, Khan SA et al (1993) 4-morpholinecarboximidine-N-1-adamantyl-N’-cyclohexylhydrochloride (U-37883A): pharmacological characterization of a novel antagonist of vascular ATP-sensitive K+ channel openers. J Pharmacol Exp Ther 266:655–665PubMed
83.
Zurück zum Zitat Humphrey SJ, Smith MP, Cimini MG et al (1996) Cardiovascular effects of the K-ATP channel blocker U-37883A and structurally related morpholinoguanidines. Methods Find Exp Clin Pharmacol 18:247–260PubMed Humphrey SJ, Smith MP, Cimini MG et al (1996) Cardiovascular effects of the K-ATP channel blocker U-37883A and structurally related morpholinoguanidines. Methods Find Exp Clin Pharmacol 18:247–260PubMed
86.
Zurück zum Zitat Muiesan G, Fariello R, Muiesan ML, Christensen OE (1985) Effect of pinacidil on blood pressure, plasma catecholamines and plasma renin activity in essential hypertension. Eur J Clin Pharmacol 28:495–499CrossRefPubMed Muiesan G, Fariello R, Muiesan ML, Christensen OE (1985) Effect of pinacidil on blood pressure, plasma catecholamines and plasma renin activity in essential hypertension. Eur J Clin Pharmacol 28:495–499CrossRefPubMed
88.
Zurück zum Zitat D’Arcy V, Laher M, McCoy D et al (1985) Pinacidil, a new vasodilator, in the treatment of mild to moderate essential hypertension. Eur J Clin Pharmacol 28:347–349CrossRefPubMed D’Arcy V, Laher M, McCoy D et al (1985) Pinacidil, a new vasodilator, in the treatment of mild to moderate essential hypertension. Eur J Clin Pharmacol 28:347–349CrossRefPubMed
89.
Zurück zum Zitat Zachariah PK, Sheps SG, Schirger A et al (1986) Antihypertensive efficacy of pinacidil--automatic ambulatory blood pressure monitoring. Eur J Clin Pharmacol 31:133–141CrossRefPubMed Zachariah PK, Sheps SG, Schirger A et al (1986) Antihypertensive efficacy of pinacidil--automatic ambulatory blood pressure monitoring. Eur J Clin Pharmacol 31:133–141CrossRefPubMed
90.
Zurück zum Zitat Sterndorff B, Johansen P (1988) The antihypertensive effect of pinacidil versus prazosin in mild to moderate hypertensive patients seen in general practice. Acta Med Scand 224:329–336CrossRefPubMed Sterndorff B, Johansen P (1988) The antihypertensive effect of pinacidil versus prazosin in mild to moderate hypertensive patients seen in general practice. Acta Med Scand 224:329–336CrossRefPubMed
91.
Zurück zum Zitat Goldberg MR (1988) Clinical pharmacology of pinacidil, a prototype for drugs that affect potassium channels. J Cardiovasc Pharmacol 12(Suppl 2):S41–S47CrossRefPubMed Goldberg MR (1988) Clinical pharmacology of pinacidil, a prototype for drugs that affect potassium channels. J Cardiovasc Pharmacol 12(Suppl 2):S41–S47CrossRefPubMed
92.
Zurück zum Zitat Camm AJ, Maltz MB (1989) A controlled single-dose study of the efficacy, dose response and duration of action of nicorandil in angina pectoris. Am J Cardiol 63:61J–65JCrossRefPubMed Camm AJ, Maltz MB (1989) A controlled single-dose study of the efficacy, dose response and duration of action of nicorandil in angina pectoris. Am J Cardiol 63:61J–65JCrossRefPubMed
93.
Zurück zum Zitat Raftery EB, Lahiri A, Hughes LO, Rose EL (1993) A double-blind comparison of a beta-blocker and a potassium channel opener in exercise induced angina. Eur heart J 14(Suppl B):35–39CrossRefPubMed Raftery EB, Lahiri A, Hughes LO, Rose EL (1993) A double-blind comparison of a beta-blocker and a potassium channel opener in exercise induced angina. Eur heart J 14(Suppl B):35–39CrossRefPubMed
94.
Zurück zum Zitat Roland E (1993) Safety profile of an anti-anginal agent with potassium channel opening activity: an overview. Eur heart J 14(Suppl B):48–52CrossRefPubMed Roland E (1993) Safety profile of an anti-anginal agent with potassium channel opening activity: an overview. Eur heart J 14(Suppl B):48–52CrossRefPubMed
95.
Zurück zum Zitat Wolf DL, Ferry JJ, Hearron AE et al (1993) The haemodynamic effects and pharmacokinetics of intravenous nicorandil in healthy volunteers. Eur J Clin Pharmacol 44:27–33CrossRefPubMed Wolf DL, Ferry JJ, Hearron AE et al (1993) The haemodynamic effects and pharmacokinetics of intravenous nicorandil in healthy volunteers. Eur J Clin Pharmacol 44:27–33CrossRefPubMed
96.
Zurück zum Zitat Witchitz S, Darmon JY (1995) Nicorandil safety in the long-term treatment of coronary heart disease. Cardiovasc Drugs Ther 9(Suppl 2):237–243CrossRefPubMed Witchitz S, Darmon JY (1995) Nicorandil safety in the long-term treatment of coronary heart disease. Cardiovasc Drugs Ther 9(Suppl 2):237–243CrossRefPubMed
97.
Zurück zum Zitat Dunn N, Bm MA, Freemantle S, et al (1999) Safety pro ® le of Nicorandil Ð prescription-event monitoring ( PEM ) study. 205:197–205 Dunn N, Bm MA, Freemantle S, et al (1999) Safety pro ® le of Nicorandil Ð prescription-event monitoring ( PEM ) study. 205:197–205
98.
Zurück zum Zitat Singer DR, Markandu ND, Miller MA et al (1989) Potassium channel stimulation in normal subjects and in patients with essential hypertension: an acute study with cromakalim (BRL 34915). J Hypertens Suppl 7:S294–S295CrossRefPubMed Singer DR, Markandu ND, Miller MA et al (1989) Potassium channel stimulation in normal subjects and in patients with essential hypertension: an acute study with cromakalim (BRL 34915). J Hypertens Suppl 7:S294–S295CrossRefPubMed
99.
Zurück zum Zitat Suzuki S, Yano K, Kusano S, Hashimoto T (1995) Antihypertensive effect of levcromakalim in patients with essential hypertension. Study by 24-h ambulatory blood pressure monitoring. Arzneimittelforschung 45:859–864PubMed Suzuki S, Yano K, Kusano S, Hashimoto T (1995) Antihypertensive effect of levcromakalim in patients with essential hypertension. Study by 24-h ambulatory blood pressure monitoring. Arzneimittelforschung 45:859–864PubMed
Metadaten
Titel
The KATP channel in migraine pathophysiology: a novel therapeutic target for migraine
verfasst von
Mohammad Al-Mahdi Al-Karagholi
Jakob Møller Hansen
Johanne Severinsen
Inger Jansen-Olesen
Messoud Ashina
Publikationsdatum
01.12.2017
Verlag
Springer Milan
Erschienen in
The Journal of Headache and Pain / Ausgabe 1/2017
Print ISSN: 1129-2369
Elektronische ISSN: 1129-2377
DOI
https://doi.org/10.1186/s10194-017-0800-8

Weitere Artikel der Ausgabe 1/2017

The Journal of Headache and Pain 1/2017 Zur Ausgabe

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.