Skip to main content
Top
Published in: The Journal of Headache and Pain 1/2019

Open Access 01-12-2019 | Migraine | Review article

Advances in genetics of migraine

Authors: Heidi G. Sutherland, Cassie L. Albury, Lyn R. Griffiths

Published in: The Journal of Headache and Pain | Issue 1/2019

Login to get access

Abstract

Background

Migraine is a complex neurovascular disorder with a strong genetic component. There are rare monogenic forms of migraine, as well as more common polygenic forms; research into the genes involved in both types has provided insights into the many contributing genetic factors. This review summarises advances that have been made in the knowledge and understanding of the genes and genetic variations implicated in migraine etiology.

Findings

Migraine is characterised into two main types, migraine without aura (MO) and migraine with aura (MA). Hemiplegic migraine is a rare monogenic MA subtype caused by mutations in three main genes - CACNA1A, ATP1A2 and SCN1A - which encode ion channel and transport proteins. Functional studies in cellular and animal models show that, in general, mutations result in impaired glutamatergic neurotransmission and cortical hyperexcitability, which make the brain more susceptible to cortical spreading depression, a phenomenon thought to coincide with aura symptoms. Variants in other genes encoding ion channels and solute carriers, or with roles in regulating neurotransmitters at neuronal synapses, or in vascular function, can also cause monogenic migraine, hemiplegic migraine and related disorders with overlapping symptoms. Next-generation sequencing will accelerate the finding of new potentially causal variants and genes, with high-throughput bioinformatics analysis methods and functional analysis pipelines important in prioritising, confirming and understanding the mechanisms of disease-causing variants.
With respect to common migraine forms, large genome-wide association studies (GWAS) have greatly expanded our knowledge of the genes involved, emphasizing the role of both neuronal and vascular pathways. Dissecting the genetic architecture of migraine leads to greater understanding of what underpins relationships between subtypes and comorbid disorders, and may have utility in diagnosis or tailoring treatments. Further work is required to identify causal polymorphisms and the mechanism of their effect, and studies of gene expression and epigenetic factors will help bridge the genetics with migraine pathophysiology.

Conclusions

The complexity of migraine disorders is mirrored by their genetic complexity. A comprehensive knowledge of the genetic factors underpinning migraine will lead to improved understanding of molecular mechanisms and pathogenesis, to enable better diagnosis and treatments for migraine sufferers.
Literature
1.
go back to reference (IHS) HCCotIHS (2018) The international classification of headache disorders, 3rd edition. Cephalalgia 38(1):1–211CrossRef (IHS) HCCotIHS (2018) The international classification of headache disorders, 3rd edition. Cephalalgia 38(1):1–211CrossRef
2.
go back to reference Goadsby PJ, Holland PR, Martins-Oliveira M, Hoffmann J, Schankin C, Akerman S (2017) Pathophysiology of migraine: a disorder of sensory processing. Physiol Rev 97(2):553–622PubMedPubMedCentralCrossRef Goadsby PJ, Holland PR, Martins-Oliveira M, Hoffmann J, Schankin C, Akerman S (2017) Pathophysiology of migraine: a disorder of sensory processing. Physiol Rev 97(2):553–622PubMedPubMedCentralCrossRef
3.
go back to reference 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(Suppl 1):S44–S53PubMedCrossRef 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(Suppl 1):S44–S53PubMedCrossRef
4.
go back to reference 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):65–80PubMedCrossRef 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):65–80PubMedCrossRef
5.
go back to reference Messlinger K, Fischer MJ, Lennerz JK (2011) Neuropeptide effects in the trigeminal system: pathophysiology and clinical relevance in migraine. Keio J Med 60(3):82–89PubMedCrossRef Messlinger K, Fischer MJ, Lennerz JK (2011) Neuropeptide effects in the trigeminal system: pathophysiology and clinical relevance in migraine. Keio J Med 60(3):82–89PubMedCrossRef
6.
go back to reference Levy D (2012) Endogenous mechanisms underlying the activation and sensitization of meningeal nociceptors: the role of immuno-vascular interactions and cortical spreading depression. Curr Pain Headache Rep 16(3):270–277PubMedCrossRef Levy D (2012) Endogenous mechanisms underlying the activation and sensitization of meningeal nociceptors: the role of immuno-vascular interactions and cortical spreading depression. Curr Pain Headache Rep 16(3):270–277PubMedCrossRef
8.
9.
go back to reference Kramer DR, Fujii T, Ohiorhenuan I, Liu CY (2016) Cortical spreading depolarization: pathophysiology, implications, and future directions. J Clin Neurosci 24:22–27PubMedCrossRef Kramer DR, Fujii T, Ohiorhenuan I, Liu CY (2016) Cortical spreading depolarization: pathophysiology, implications, and future directions. J Clin Neurosci 24:22–27PubMedCrossRef
10.
go back to reference Goadsby PJ (2001) Migraine, aura, and cortical spreading depression: why are we still talking about it? Ann Neurol 49(1):4–6PubMedCrossRef Goadsby PJ (2001) Migraine, aura, and cortical spreading depression: why are we still talking about it? Ann Neurol 49(1):4–6PubMedCrossRef
11.
go back to reference Bolay H, Reuter U, Dunn AK, Huang Z, Boas DA, Moskowitz MA (2002) Intrinsic brain activity triggers trigeminal meningeal afferents in a migraine model. Nat Med 8(2):136–142PubMedCrossRef Bolay H, Reuter U, Dunn AK, Huang Z, Boas DA, Moskowitz MA (2002) Intrinsic brain activity triggers trigeminal meningeal afferents in a migraine model. Nat Med 8(2):136–142PubMedCrossRef
12.
go back to reference Zhang X, Levy D, Kainz V, Noseda R, Jakubowski M, Burstein R (2011) Activation of central trigeminovascular neurons by cortical spreading depression. Ann Neurol 69(5):855–865PubMedPubMedCentralCrossRef Zhang X, Levy D, Kainz V, Noseda R, Jakubowski M, Burstein R (2011) Activation of central trigeminovascular neurons by cortical spreading depression. Ann Neurol 69(5):855–865PubMedPubMedCentralCrossRef
13.
go back to reference Karatas H, Erdener SE, Gursoy-Ozdemir Y, Lule S, Eren-Kocak E, Sen ZD et al (2013) Spreading depression triggers headache by activating neuronal Panx1 channels. Science 339(6123):1092–1095PubMedCrossRef Karatas H, Erdener SE, Gursoy-Ozdemir Y, Lule S, Eren-Kocak E, Sen ZD et al (2013) Spreading depression triggers headache by activating neuronal Panx1 channels. Science 339(6123):1092–1095PubMedCrossRef
15.
go back to reference Stankewitz A, Aderjan D, Eippert F, May A (2011) Trigeminal nociceptive transmission in migraineurs predicts migraine attacks. J Neurosci 31(6):1937–1943PubMedCrossRefPubMedCentral Stankewitz A, Aderjan D, Eippert F, May A (2011) Trigeminal nociceptive transmission in migraineurs predicts migraine attacks. J Neurosci 31(6):1937–1943PubMedCrossRefPubMedCentral
16.
go back to reference Lang E, Kaltenhauser M, Neundorfer B, Seidler S (2004) Hyperexcitability of the primary somatosensory cortex in migraine--a magnetoencephalographic study. Brain 127(Pt 11):2459–2469PubMedCrossRef Lang E, Kaltenhauser M, Neundorfer B, Seidler S (2004) Hyperexcitability of the primary somatosensory cortex in migraine--a magnetoencephalographic study. Brain 127(Pt 11):2459–2469PubMedCrossRef
17.
go back to reference Aurora SK, Barrodale PM, Tipton RL, Khodavirdi A (2007) Brainstem dysfunction in chronic migraine as evidenced by neurophysiological and positron emission tomography studies. Headache 47(7):996–1003 discussion 4-7PubMedCrossRef Aurora SK, Barrodale PM, Tipton RL, Khodavirdi A (2007) Brainstem dysfunction in chronic migraine as evidenced by neurophysiological and positron emission tomography studies. Headache 47(7):996–1003 discussion 4-7PubMedCrossRef
18.
go back to reference Vecchia D, Pietrobon D (2012) Migraine: a disorder of brain excitatory-inhibitory balance? Trends Neurosci 35(8):507–520PubMedCrossRef Vecchia D, Pietrobon D (2012) Migraine: a disorder of brain excitatory-inhibitory balance? Trends Neurosci 35(8):507–520PubMedCrossRef
19.
go back to reference Honkasalo ML, Kaprio J, Winter T, Heikkila K, Sillanpaa M, Koskenvuo M (1995) Migraine and concomitant symptoms among 8167 adult twin pairs. Headache 35(2):70–78PubMedCrossRef Honkasalo ML, Kaprio J, Winter T, Heikkila K, Sillanpaa M, Koskenvuo M (1995) Migraine and concomitant symptoms among 8167 adult twin pairs. Headache 35(2):70–78PubMedCrossRef
20.
go back to reference Mulder EJ, Van Baal C, Gaist D, Kallela M, Kaprio J, Svensson DA et al (2003) Genetic and environmental influences on migraine: a twin study across six countries. Twin Res 6(5):422–431PubMedCrossRef Mulder EJ, Van Baal C, Gaist D, Kallela M, Kaprio J, Svensson DA et al (2003) Genetic and environmental influences on migraine: a twin study across six countries. Twin Res 6(5):422–431PubMedCrossRef
21.
go back to reference Polderman TJ, Benyamin B, de Leeuw CA, Sullivan PF, van Bochoven A, Visscher PM et al (2015) Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat Genet 47(7):702–709PubMedCrossRef Polderman TJ, Benyamin B, de Leeuw CA, Sullivan PF, van Bochoven A, Visscher PM et al (2015) Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat Genet 47(7):702–709PubMedCrossRef
22.
go back to reference Pelzer N, Stam AH, Haan J, Ferrari MD, Terwindt GM (2013) Familial and sporadic hemiplegic migraine: diagnosis and treatment. Curr Treat Options Neurol 15(1):13–27PubMedCrossRef Pelzer N, Stam AH, Haan J, Ferrari MD, Terwindt GM (2013) Familial and sporadic hemiplegic migraine: diagnosis and treatment. Curr Treat Options Neurol 15(1):13–27PubMedCrossRef
23.
go back to reference Russell MB, Ducros A (2011) Sporadic and familial hemiplegic migraine: pathophysiological mechanisms, clinical characteristics, diagnosis, and management. Lancet Neurol 10(5):457–470PubMedCrossRef Russell MB, Ducros A (2011) Sporadic and familial hemiplegic migraine: pathophysiological mechanisms, clinical characteristics, diagnosis, and management. Lancet Neurol 10(5):457–470PubMedCrossRef
24.
go back to reference Kors EE, Terwindt GM, Vermeulen FL, Fitzsimons RB, Jardine PE, Heywood P et al (2001) Delayed cerebral edema and fatal coma after minor head trauma: role of the CACNA1A calcium channel subunit gene and relationship with familial hemiplegic migraine. Ann Neurol 49(6):753–760PubMedCrossRef Kors EE, Terwindt GM, Vermeulen FL, Fitzsimons RB, Jardine PE, Heywood P et al (2001) Delayed cerebral edema and fatal coma after minor head trauma: role of the CACNA1A calcium channel subunit gene and relationship with familial hemiplegic migraine. Ann Neurol 49(6):753–760PubMedCrossRef
25.
go back to reference Lykke Thomsen L, Kirchmann Eriksen M, Faerch Romer S, Andersen I, Ostergaard E, Keiding N et al (2002) An epidemiological survey of hemiplegic migraine. Cephalalgia 22(5):361–375PubMedCrossRef Lykke Thomsen L, Kirchmann Eriksen M, Faerch Romer S, Andersen I, Ostergaard E, Keiding N et al (2002) An epidemiological survey of hemiplegic migraine. Cephalalgia 22(5):361–375PubMedCrossRef
26.
go back to reference Barros J, Ruano L, Domingos J, Tuna A, Damasio J, Alonso I et al (2014) The prevalence of familial hemiplegic migraine with cerebellar ataxia and spinocerebellar ataxia type 6 in Portugal. Headache 54(5):911–915PubMedCrossRef Barros J, Ruano L, Domingos J, Tuna A, Damasio J, Alonso I et al (2014) The prevalence of familial hemiplegic migraine with cerebellar ataxia and spinocerebellar ataxia type 6 in Portugal. Headache 54(5):911–915PubMedCrossRef
27.
go back to reference Terwindt GM, Ophoff RA, Haan J, Vergouwe MN, van Eijk R, Frants RR et al (1998) Variable clinical expression of mutations in the P/Q-type calcium channel gene in familial hemiplegic migraine. Dutch Migraine Genetics Research Group. Neurology 50(4):1105–1110PubMedCrossRef Terwindt GM, Ophoff RA, Haan J, Vergouwe MN, van Eijk R, Frants RR et al (1998) Variable clinical expression of mutations in the P/Q-type calcium channel gene in familial hemiplegic migraine. Dutch Migraine Genetics Research Group. Neurology 50(4):1105–1110PubMedCrossRef
29.
go back to reference Kors EE, Haan J, Giffin NJ, Pazdera L, Schnittger C, Lennox GG et al (2003) Expanding the phenotypic spectrum of the CACNA1A gene T666M mutation: a description of 5 families with familial hemiplegic migraine. Arch Neurol 60(5):684–688PubMedCrossRef Kors EE, Haan J, Giffin NJ, Pazdera L, Schnittger C, Lennox GG et al (2003) Expanding the phenotypic spectrum of the CACNA1A gene T666M mutation: a description of 5 families with familial hemiplegic migraine. Arch Neurol 60(5):684–688PubMedCrossRef
30.
go back to reference Hiekkala ME, Vuola P, Artto V, Happola P, Happola E, Vepsalainen S et al (2018) The contribution of CACNA1A, ATP1A2 and SCN1A mutations in hemiplegic migraine: a clinical and genetic study in Finnish migraine families. Cephalalgia 38(12):1849–1863PubMedCrossRef Hiekkala ME, Vuola P, Artto V, Happola P, Happola E, Vepsalainen S et al (2018) The contribution of CACNA1A, ATP1A2 and SCN1A mutations in hemiplegic migraine: a clinical and genetic study in Finnish migraine families. Cephalalgia 38(12):1849–1863PubMedCrossRef
32.
go back to reference Ophoff RA, Terwindt GM, Vergouwe MN, van Eijk R, Oefner PJ, Hoffman SM et al (1996) Familial hemiplegic migraine and episodic ataxia type-2 are caused by mutations in the Ca2+ channel gene CACNL1A4. Cell 87(3):543–552PubMedCrossRef Ophoff RA, Terwindt GM, Vergouwe MN, van Eijk R, Oefner PJ, Hoffman SM et al (1996) Familial hemiplegic migraine and episodic ataxia type-2 are caused by mutations in the Ca2+ channel gene CACNL1A4. Cell 87(3):543–552PubMedCrossRef
33.
go back to reference Catterall WA (1998) Structure and function of neuronal Ca2+ channels and their role in neurotransmitter release. Cell Calcium 24(5–6):307–323PubMedCrossRef Catterall WA (1998) Structure and function of neuronal Ca2+ channels and their role in neurotransmitter release. Cell Calcium 24(5–6):307–323PubMedCrossRef
34.
go back to reference Labrum RW, Rajakulendran S, Graves TD, Eunson LH, Bevan R, Sweeney MG et al (2009) Large scale calcium channel gene rearrangements in episodic ataxia and hemiplegic migraine: implications for diagnostic testing. J Med Genet 46(11):786–791PubMedCrossRef Labrum RW, Rajakulendran S, Graves TD, Eunson LH, Bevan R, Sweeney MG et al (2009) Large scale calcium channel gene rearrangements in episodic ataxia and hemiplegic migraine: implications for diagnostic testing. J Med Genet 46(11):786–791PubMedCrossRef
35.
go back to reference Grieco GS, Gagliardi S, Ricca I, Pansarasa O, Neri M, Gualandi F et al (2018) New CACNA1A deletions are associated to migraine phenotypes. J Headache Pain 19(1):75PubMedPubMedCentralCrossRef Grieco GS, Gagliardi S, Ricca I, Pansarasa O, Neri M, Gualandi F et al (2018) New CACNA1A deletions are associated to migraine phenotypes. J Headache Pain 19(1):75PubMedPubMedCentralCrossRef
36.
go back to reference Ducros A, Denier C, Joutel A, Cecillon M, Lescoat C, Vahedi K et al (2001) The clinical spectrum of familial hemiplegic migraine associated with mutations in a neuronal calcium channel. N Engl J Med 345(1):17–24PubMedCrossRef Ducros A, Denier C, Joutel A, Cecillon M, Lescoat C, Vahedi K et al (2001) The clinical spectrum of familial hemiplegic migraine associated with mutations in a neuronal calcium channel. N Engl J Med 345(1):17–24PubMedCrossRef
37.
go back to reference Di Lorenzo C, Grieco GS, Santorelli FM (2012) Migraine headache: a review of the molecular genetics of a common disorder. J Headache Pain 13(7):571–580PubMedPubMedCentralCrossRef Di Lorenzo C, Grieco GS, Santorelli FM (2012) Migraine headache: a review of the molecular genetics of a common disorder. J Headache Pain 13(7):571–580PubMedPubMedCentralCrossRef
38.
go back to reference Tottene A, Fellin T, Pagnutti S, Luvisetto S, Striessnig J, Fletcher C et al (2002) Familial hemiplegic migraine mutations increase ca (2+) influx through single human CaV2.1 channels and decrease maximal CaV2.1 current density in neurons. Proc Natl Acad Sci U S A 99(20):13284–13289PubMedPubMedCentralCrossRef Tottene A, Fellin T, Pagnutti S, Luvisetto S, Striessnig J, Fletcher C et al (2002) Familial hemiplegic migraine mutations increase ca (2+) influx through single human CaV2.1 channels and decrease maximal CaV2.1 current density in neurons. Proc Natl Acad Sci U S A 99(20):13284–13289PubMedPubMedCentralCrossRef
39.
go back to reference Tournier-Lasserve E (1999) CACNA1A mutations: hemiplegic migraine, episodic ataxia type 2, and the others. Neurology 53(1):3–4PubMedCrossRef Tournier-Lasserve E (1999) CACNA1A mutations: hemiplegic migraine, episodic ataxia type 2, and the others. Neurology 53(1):3–4PubMedCrossRef
40.
go back to reference de Vries B, Frants RR, Ferrari MD, van den Maagdenberg AM (2009) Molecular genetics of migraine. Hum Genet 126(1):115–132PubMedCrossRef de Vries B, Frants RR, Ferrari MD, van den Maagdenberg AM (2009) Molecular genetics of migraine. Hum Genet 126(1):115–132PubMedCrossRef
41.
go back to reference Pelzer N, Haan J, Stam AH, Vijfhuizen LS, Koelewijn SC, Smagge A et al (2018) Clinical spectrum of hemiplegic migraine and chances of finding a pathogenic mutation. Neurology 90(7):e575–ee82PubMedCrossRef Pelzer N, Haan J, Stam AH, Vijfhuizen LS, Koelewijn SC, Smagge A et al (2018) Clinical spectrum of hemiplegic migraine and chances of finding a pathogenic mutation. Neurology 90(7):e575–ee82PubMedCrossRef
42.
go back to reference van den Maagdenberg AM, Pietrobon D, Pizzorusso T, Kaja S, Broos LA, Cesetti T et al (2004) A Cacna1a knockin migraine mouse model with increased susceptibility to cortical spreading depression. Neuron 41(5):701–710PubMedCrossRef van den Maagdenberg AM, Pietrobon D, Pizzorusso T, Kaja S, Broos LA, Cesetti T et al (2004) A Cacna1a knockin migraine mouse model with increased susceptibility to cortical spreading depression. Neuron 41(5):701–710PubMedCrossRef
43.
go back to reference Eikermann-Haerter K, Dilekoz E, Kudo C, Savitz SI, Waeber C, Baum MJ et al (2009) Genetic and hormonal factors modulate spreading depression and transient hemiparesis in mouse models of familial hemiplegic migraine type 1. J Clin Invest 119(1):99–109PubMed Eikermann-Haerter K, Dilekoz E, Kudo C, Savitz SI, Waeber C, Baum MJ et al (2009) Genetic and hormonal factors modulate spreading depression and transient hemiparesis in mouse models of familial hemiplegic migraine type 1. J Clin Invest 119(1):99–109PubMed
44.
go back to reference Eroli F, Vilotti S, van den Maagdenberg A, Nistri A (2017) Hyperpolarization-activated current Ih in mouse trigeminal sensory neurons in a transgenic mouse model of familial hemiplegic migraine type-1. Neuroscience 351:47–64PubMedCrossRef Eroli F, Vilotti S, van den Maagdenberg A, Nistri A (2017) Hyperpolarization-activated current Ih in mouse trigeminal sensory neurons in a transgenic mouse model of familial hemiplegic migraine type-1. Neuroscience 351:47–64PubMedCrossRef
45.
go back to reference Tottene A, Conti R, Fabbro A, Vecchia D, Shapovalova M, Santello M et al (2009) Enhanced excitatory transmission at cortical synapses as the basis for facilitated spreading depression in ca(v)2.1 knockin migraine mice. Neuron 61(5):762–773PubMedCrossRef Tottene A, Conti R, Fabbro A, Vecchia D, Shapovalova M, Santello M et al (2009) Enhanced excitatory transmission at cortical synapses as the basis for facilitated spreading depression in ca(v)2.1 knockin migraine mice. Neuron 61(5):762–773PubMedCrossRef
46.
go back to reference Hullugundi SK, Ansuini A, Ferrari MD, van den Maagdenberg AM, Nistri A (2014) A hyperexcitability phenotype in mouse trigeminal sensory neurons expressing the R192Q Cacna1a missense mutation of familial hemiplegic migraine type-1. Neuroscience 266:244–254PubMedCrossRef Hullugundi SK, Ansuini A, Ferrari MD, van den Maagdenberg AM, Nistri A (2014) A hyperexcitability phenotype in mouse trigeminal sensory neurons expressing the R192Q Cacna1a missense mutation of familial hemiplegic migraine type-1. Neuroscience 266:244–254PubMedCrossRef
47.
go back to reference Marchenkova A, van den Maagdenberg AM, Nistri A (2016) Loss of inhibition by brain natriuretic peptide over P2X3 receptors contributes to enhanced spike firing of trigeminal ganglion neurons in a mouse model of familial hemiplegic migraine type-1. Neuroscience 331:197–205PubMedCrossRef Marchenkova A, van den Maagdenberg AM, Nistri A (2016) Loss of inhibition by brain natriuretic peptide over P2X3 receptors contributes to enhanced spike firing of trigeminal ganglion neurons in a mouse model of familial hemiplegic migraine type-1. Neuroscience 331:197–205PubMedCrossRef
48.
go back to reference Khennouf L, Gesslein B, Lind BL, van den Maagdenberg AM, Lauritzen M (2016) Activity-dependent calcium, oxygen, and vascular responses in a mouse model of familial hemiplegic migraine type 1. Ann Neurol 80(2):219–232PubMedCrossRef Khennouf L, Gesslein B, Lind BL, van den Maagdenberg AM, Lauritzen M (2016) Activity-dependent calcium, oxygen, and vascular responses in a mouse model of familial hemiplegic migraine type 1. Ann Neurol 80(2):219–232PubMedCrossRef
49.
go back to reference Chanda ML, Tuttle AH, Baran I, Atlin C, Guindi D, Hathaway G et al (2013) Behavioral evidence for photophobia and stress-related ipsilateral head pain in transgenic Cacna1a mutant mice. Pain 154(8):1254–1262PubMedCrossRef Chanda ML, Tuttle AH, Baran I, Atlin C, Guindi D, Hathaway G et al (2013) Behavioral evidence for photophobia and stress-related ipsilateral head pain in transgenic Cacna1a mutant mice. Pain 154(8):1254–1262PubMedCrossRef
50.
go back to reference Chen SP, Tolner EA, Eikermann-Haerter K (2016) Animal models of monogenic migraine. Cephalalgia 36(7):704–721PubMedCrossRef Chen SP, Tolner EA, Eikermann-Haerter K (2016) Animal models of monogenic migraine. Cephalalgia 36(7):704–721PubMedCrossRef
51.
go back to reference Brusich DJ, Spring AM, James TD, Yeates CJ, Helms TH, Frank CA (2018) Drosophila CaV2 channels harboring human migraine mutations cause synapse hyperexcitability that can be suppressed by inhibition of a Ca2+ store release pathway. PLoS Genet 14(8):e1007577PubMedPubMedCentralCrossRef Brusich DJ, Spring AM, James TD, Yeates CJ, Helms TH, Frank CA (2018) Drosophila CaV2 channels harboring human migraine mutations cause synapse hyperexcitability that can be suppressed by inhibition of a Ca2+ store release pathway. PLoS Genet 14(8):e1007577PubMedPubMedCentralCrossRef
52.
go back to reference Pereira Mda C, Morais S, Sequeiros J, Alonso I (2016) Large-Scale Functional RNAi Screen in C. elegans Identifies TGF-beta and Notch Signaling Pathways as Modifiers of CACNA1A. ASN Neuro 8(2):1-10CrossRef Pereira Mda C, Morais S, Sequeiros J, Alonso I (2016) Large-Scale Functional RNAi Screen in C. elegans Identifies TGF-beta and Notch Signaling Pathways as Modifiers of CACNA1A. ASN Neuro 8(2):1-10CrossRef
53.
go back to reference Gormley P, Anttila V, Winsvold BS, Palta P, Esko T, Pers TH et al (2016) Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat Genet 48(8):856–866PubMedPubMedCentralCrossRef Gormley P, Anttila V, Winsvold BS, Palta P, Esko T, Pers TH et al (2016) Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat Genet 48(8):856–866PubMedPubMedCentralCrossRef
54.
go back to reference Chabriat H, Joutel A, Dichgans M, Tournier-Lasserve E, Bousser MG (2009) Cadasil. Lancet Neurol 8(7):643–653PubMedCrossRef Chabriat H, Joutel A, Dichgans M, Tournier-Lasserve E, Bousser MG (2009) Cadasil. Lancet Neurol 8(7):643–653PubMedCrossRef
55.
go back to reference Zhuchenko O, Bailey J, Bonnen P, Ashizawa T, Stockton DW, Amos C et al (1997) Autosomal dominant cerebellar ataxia (SCA6) associated with small polyglutamine expansions in the alpha 1A-voltage-dependent calcium channel. Nat Genet 15(1):62–69CrossRefPubMed Zhuchenko O, Bailey J, Bonnen P, Ashizawa T, Stockton DW, Amos C et al (1997) Autosomal dominant cerebellar ataxia (SCA6) associated with small polyglutamine expansions in the alpha 1A-voltage-dependent calcium channel. Nat Genet 15(1):62–69CrossRefPubMed
56.
go back to reference Blumenfeld AE, Victorio MC, Berenson FR (2016) Complicated migraines. Semin Pediatr Neurol 23(1):18–22PubMedCrossRef Blumenfeld AE, Victorio MC, Berenson FR (2016) Complicated migraines. Semin Pediatr Neurol 23(1):18–22PubMedCrossRef
57.
go back to reference Jen J, Kim GW, Baloh RW (2004) Clinical spectrum of episodic ataxia type 2. Neurology 62(1):17–22PubMedCrossRef Jen J, Kim GW, Baloh RW (2004) Clinical spectrum of episodic ataxia type 2. Neurology 62(1):17–22PubMedCrossRef
58.
go back to reference Sinke RJ, Ippel EF, Diepstraten CM, Beemer FA, Wokke JH, van Hilten BJ et al (2001) Clinical and molecular correlations in spinocerebellar ataxia type 6: a study of 24 Dutch families. Arch Neurol 58(11):1839–1844PubMedCrossRef Sinke RJ, Ippel EF, Diepstraten CM, Beemer FA, Wokke JH, van Hilten BJ et al (2001) Clinical and molecular correlations in spinocerebellar ataxia type 6: a study of 24 Dutch families. Arch Neurol 58(11):1839–1844PubMedCrossRef
59.
go back to reference Mantuano E, Romano S, Veneziano L, Gellera C, Castellotti B, Caimi S et al (2010) Identification of novel and recurrent CACNA1A gene mutations in fifteen patients with episodic ataxia type 2. J Neurol Sci 291(1–2):30–36PubMedCrossRef Mantuano E, Romano S, Veneziano L, Gellera C, Castellotti B, Caimi S et al (2010) Identification of novel and recurrent CACNA1A gene mutations in fifteen patients with episodic ataxia type 2. J Neurol Sci 291(1–2):30–36PubMedCrossRef
60.
go back to reference Watase K, Barrett CF, Miyazaki T, Ishiguro T, Ishikawa K, Hu Y et al (2008) Spinocerebellar ataxia type 6 knockin mice develop a progressive neuronal dysfunction with age-dependent accumulation of mutant CaV2.1 channels. Proc Natl Acad Sci U S A 105(33):11987–11992PubMedPubMedCentralCrossRef Watase K, Barrett CF, Miyazaki T, Ishiguro T, Ishikawa K, Hu Y et al (2008) Spinocerebellar ataxia type 6 knockin mice develop a progressive neuronal dysfunction with age-dependent accumulation of mutant CaV2.1 channels. Proc Natl Acad Sci U S A 105(33):11987–11992PubMedPubMedCentralCrossRef
61.
go back to reference De Fusco M, Marconi R, Silvestri L, Atorino L, Rampoldi L, Morgante L et al (2003) Haploinsufficiency of ATP1A2 encoding the Na+/K+ pump alpha2 subunit associated with familial hemiplegic migraine type 2. Nat Genet 33(2):192–196PubMedCrossRef De Fusco M, Marconi R, Silvestri L, Atorino L, Rampoldi L, Morgante L et al (2003) Haploinsufficiency of ATP1A2 encoding the Na+/K+ pump alpha2 subunit associated with familial hemiplegic migraine type 2. Nat Genet 33(2):192–196PubMedCrossRef
62.
go back to reference Friedrich T, Tavraz NN, Junghans C (2016) ATP1A2 mutations in migraine: seeing through the facets of an ion pump onto the neurobiology of disease. Front Physiol 7:239PubMedPubMedCentral Friedrich T, Tavraz NN, Junghans C (2016) ATP1A2 mutations in migraine: seeing through the facets of an ion pump onto the neurobiology of disease. Front Physiol 7:239PubMedPubMedCentral
63.
go back to reference Benarroch EE (2010) Glutamate transporters: diversity, function, and involvement in neurologic disease. Neurology 74(3):259–264PubMedCrossRef Benarroch EE (2010) Glutamate transporters: diversity, function, and involvement in neurologic disease. Neurology 74(3):259–264PubMedCrossRef
64.
go back to reference Jurkat-Rott K, Freilinger T, Dreier JP, Herzog J, Gobel H, Petzold GC et al (2004) Variability of familial hemiplegic migraine with novel A1A2 Na+/K+-ATPase variants. Neurology 62(10):1857–1861PubMedCrossRef Jurkat-Rott K, Freilinger T, Dreier JP, Herzog J, Gobel H, Petzold GC et al (2004) Variability of familial hemiplegic migraine with novel A1A2 Na+/K+-ATPase variants. Neurology 62(10):1857–1861PubMedCrossRef
65.
go back to reference Bassi MT, Bresolin N, Tonelli A, Nazos K, Crippa F, Baschirotto C et al (2004) A novel mutation in the ATP1A2 gene causes alternating hemiplegia of childhood. J Med Genet 41(8):621–628PubMedPubMedCentralCrossRef Bassi MT, Bresolin N, Tonelli A, Nazos K, Crippa F, Baschirotto C et al (2004) A novel mutation in the ATP1A2 gene causes alternating hemiplegia of childhood. J Med Genet 41(8):621–628PubMedPubMedCentralCrossRef
66.
go back to reference Deprez L, Weckhuysen S, Peeters K, Deconinck T, Claeys KG, Claes LR et al (2008) Epilepsy as part of the phenotype associated with ATP1A2 mutations. Epilepsia 49(3):500–508PubMedCrossRef Deprez L, Weckhuysen S, Peeters K, Deconinck T, Claeys KG, Claes LR et al (2008) Epilepsy as part of the phenotype associated with ATP1A2 mutations. Epilepsia 49(3):500–508PubMedCrossRef
67.
go back to reference Al-Bulushi B, Al-Hashem A, Tabarki B (2014) A wide clinical phenotype spectrum in patients with ATP1A2 mutations. J Child Neurol 29(2):265–268PubMedCrossRef Al-Bulushi B, Al-Hashem A, Tabarki B (2014) A wide clinical phenotype spectrum in patients with ATP1A2 mutations. J Child Neurol 29(2):265–268PubMedCrossRef
68.
go back to reference Vanmolkot KR, Stroink H, Koenderink JB, Kors EE, van den Heuvel JJ, van den Boogerd EH et al (2006) Severe episodic neurological deficits and permanent mental retardation in a child with a novel FHM2 ATP1A2 mutation. Ann Neurol 59(2):310–314PubMedCrossRef Vanmolkot KR, Stroink H, Koenderink JB, Kors EE, van den Heuvel JJ, van den Boogerd EH et al (2006) Severe episodic neurological deficits and permanent mental retardation in a child with a novel FHM2 ATP1A2 mutation. Ann Neurol 59(2):310–314PubMedCrossRef
69.
go back to reference Roth C, Freilinger T, Kirovski G, Dunkel J, Shah Y, Wilken B et al (2014) Clinical spectrum in three families with familial hemiplegic migraine type 2 including a novel mutation in the ATP1A2 gene. Cephalalgia 34(3):183–190PubMedCrossRef Roth C, Freilinger T, Kirovski G, Dunkel J, Shah Y, Wilken B et al (2014) Clinical spectrum in three families with familial hemiplegic migraine type 2 including a novel mutation in the ATP1A2 gene. Cephalalgia 34(3):183–190PubMedCrossRef
70.
go back to reference Sampedro Castaneda M, Zanoteli E, Scalco RS, Scaramuzzi V, Marques Caldas V, Conti Reed U et al (2018) A novel ATP1A2 mutation in a patient with hypokalaemic periodic paralysis and CNS symptoms. Brain 141(12):3308–3318PubMedPubMedCentralCrossRef Sampedro Castaneda M, Zanoteli E, Scalco RS, Scaramuzzi V, Marques Caldas V, Conti Reed U et al (2018) A novel ATP1A2 mutation in a patient with hypokalaemic periodic paralysis and CNS symptoms. Brain 141(12):3308–3318PubMedPubMedCentralCrossRef
71.
go back to reference Pelzer N, Blom DE, Stam AH, Vijfhuizen LS, Hageman A, van Vliet JA et al (2017) Recurrent coma and fever in familial hemiplegic migraine type 2. A prospective 15-year follow-up of a large family with a novel ATP1A2 mutation. Cephalalgia 37(8):737–755PubMedCrossRef Pelzer N, Blom DE, Stam AH, Vijfhuizen LS, Hageman A, van Vliet JA et al (2017) Recurrent coma and fever in familial hemiplegic migraine type 2. A prospective 15-year follow-up of a large family with a novel ATP1A2 mutation. Cephalalgia 37(8):737–755PubMedCrossRef
72.
go back to reference Carreno O, Corominas R, Serra SA, Sintas C, Fernandez-Castillo N, Vila-Pueyo M et al (2013) Screening of CACNA1A and ATP1A2 genes in hemiplegic migraine: clinical, genetic, and functional studies. Mol Genet Genomic Med 1(4):206–222PubMedPubMedCentralCrossRef Carreno O, Corominas R, Serra SA, Sintas C, Fernandez-Castillo N, Vila-Pueyo M et al (2013) Screening of CACNA1A and ATP1A2 genes in hemiplegic migraine: clinical, genetic, and functional studies. Mol Genet Genomic Med 1(4):206–222PubMedPubMedCentralCrossRef
73.
go back to reference Riant F, De Fusco M, Aridon P, Ducros A, Ploton C, Marchelli F et al (2005) ATP1A2 mutations in 11 families with familial hemiplegic migraine. Hum Mutat 26(3):281PubMedCrossRef Riant F, De Fusco M, Aridon P, Ducros A, Ploton C, Marchelli F et al (2005) ATP1A2 mutations in 11 families with familial hemiplegic migraine. Hum Mutat 26(3):281PubMedCrossRef
74.
go back to reference Riant F, Ducros A, Ploton C, Barbance C, Depienne C, Tournier-Lasserve E (2010) De novo mutations in ATP1A2 and CACNA1A are frequent in early-onset sporadic hemiplegic migraine. Neurology 75(11):967–972PubMedCrossRef Riant F, Ducros A, Ploton C, Barbance C, Depienne C, Tournier-Lasserve E (2010) De novo mutations in ATP1A2 and CACNA1A are frequent in early-onset sporadic hemiplegic migraine. Neurology 75(11):967–972PubMedCrossRef
75.
go back to reference Gagliardi S, Grieco GS, Gualandi F, Caniatti LM, Groppo E, Valente M et al (2017) De novo exonic duplication of ATP1A2 in Italian patient with hemiplegic migraine: a case report. J Headache Pain 18(1):63PubMedPubMedCentralCrossRef Gagliardi S, Grieco GS, Gualandi F, Caniatti LM, Groppo E, Valente M et al (2017) De novo exonic duplication of ATP1A2 in Italian patient with hemiplegic migraine: a case report. J Headache Pain 18(1):63PubMedPubMedCentralCrossRef
76.
go back to reference Segall L, Mezzetti A, Scanzano R, Gargus JJ, Purisima E, Blostein R (2005) Alterations in the alpha2 isoform of Na,K-ATPase associated with familial hemiplegic migraine type 2. Proc Natl Acad Sci U S A 102(31):11106–11111PubMedPubMedCentralCrossRef Segall L, Mezzetti A, Scanzano R, Gargus JJ, Purisima E, Blostein R (2005) Alterations in the alpha2 isoform of Na,K-ATPase associated with familial hemiplegic migraine type 2. Proc Natl Acad Sci U S A 102(31):11106–11111PubMedPubMedCentralCrossRef
77.
go back to reference Tavraz NN, Friedrich T, Durr KL, Koenderink JB, Bamberg E, Freilinger T et al (2008) Diverse functional consequences of mutations in the Na+/K+-ATPase alpha2-subunit causing familial hemiplegic migraine type 2. J Biol Chem 283(45):31097–31106PubMedPubMedCentralCrossRef Tavraz NN, Friedrich T, Durr KL, Koenderink JB, Bamberg E, Freilinger T et al (2008) Diverse functional consequences of mutations in the Na+/K+-ATPase alpha2-subunit causing familial hemiplegic migraine type 2. J Biol Chem 283(45):31097–31106PubMedPubMedCentralCrossRef
78.
go back to reference Capendeguy O, Horisberger JD (2004) Functional effects of Na+,K+-ATPase gene mutations linked to familial hemiplegic migraine. NeuroMolecular Med 6(2–3):105–116PubMedCrossRef Capendeguy O, Horisberger JD (2004) Functional effects of Na+,K+-ATPase gene mutations linked to familial hemiplegic migraine. NeuroMolecular Med 6(2–3):105–116PubMedCrossRef
79.
go back to reference Koenderink JB, Zifarelli G, Qiu LY, Schwarz W, De Pont JJ, Bamberg E et al (2005) Na,K-ATPase mutations in familial hemiplegic migraine lead to functional inactivation. Biochim Biophys Acta 1669(1):61–68PubMedCrossRef Koenderink JB, Zifarelli G, Qiu LY, Schwarz W, De Pont JJ, Bamberg E et al (2005) Na,K-ATPase mutations in familial hemiplegic migraine lead to functional inactivation. Biochim Biophys Acta 1669(1):61–68PubMedCrossRef
80.
go back to reference Tavraz NN, Durr KL, Koenderink JB, Freilinger T, Bamberg E, Dichgans M et al (2009) Impaired plasma membrane targeting or protein stability by certain ATP1A2 mutations identified in sporadic or familial hemiplegic migraine. Channels (Austin) 3(2):82–87CrossRef Tavraz NN, Durr KL, Koenderink JB, Freilinger T, Bamberg E, Dichgans M et al (2009) Impaired plasma membrane targeting or protein stability by certain ATP1A2 mutations identified in sporadic or familial hemiplegic migraine. Channels (Austin) 3(2):82–87CrossRef
81.
go back to reference Ikeda K, Onaka T, Yamakado M, Nakai J, Ishikawa TO, Taketo MM et al (2003) Degeneration of the amygdala/piriform cortex and enhanced fear/anxiety behaviors in sodium pump alpha2 subunit (Atp1a2)-deficient mice. J Neurosci 23(11):4667–4676PubMedCrossRefPubMedCentral Ikeda K, Onaka T, Yamakado M, Nakai J, Ishikawa TO, Taketo MM et al (2003) Degeneration of the amygdala/piriform cortex and enhanced fear/anxiety behaviors in sodium pump alpha2 subunit (Atp1a2)-deficient mice. J Neurosci 23(11):4667–4676PubMedCrossRefPubMedCentral
82.
83.
go back to reference Unekawa M, Ikeda K, Tomita Y, Kawakami K, Suzuki N (2018) Enhanced susceptibility to cortical spreading depression in two types of Na(+),K(+)-ATPase alpha2 subunit-deficient mice as a model of familial hemiplegic migraine 2. Cephalalgia 38(9):1515–1524PubMedCrossRef Unekawa M, Ikeda K, Tomita Y, Kawakami K, Suzuki N (2018) Enhanced susceptibility to cortical spreading depression in two types of Na(+),K(+)-ATPase alpha2 subunit-deficient mice as a model of familial hemiplegic migraine 2. Cephalalgia 38(9):1515–1524PubMedCrossRef
84.
go back to reference Leo L, Gherardini L, Barone V, De Fusco M, Pietrobon D, Pizzorusso T et al (2011) Increased susceptibility to cortical spreading depression in the mouse model of familial hemiplegic migraine type 2. PLoS Genet 7(6):e1002129PubMedPubMedCentralCrossRef Leo L, Gherardini L, Barone V, De Fusco M, Pietrobon D, Pizzorusso T et al (2011) Increased susceptibility to cortical spreading depression in the mouse model of familial hemiplegic migraine type 2. PLoS Genet 7(6):e1002129PubMedPubMedCentralCrossRef
85.
go back to reference Capuani C, Melone M, Tottene A, Bragina L, Crivellaro G, Santello M et al (2016) Defective glutamate and K+ clearance by cortical astrocytes in familial hemiplegic migraine type 2. EMBO Mol Med 8(8):967–986PubMedPubMedCentralCrossRef Capuani C, Melone M, Tottene A, Bragina L, Crivellaro G, Santello M et al (2016) Defective glutamate and K+ clearance by cortical astrocytes in familial hemiplegic migraine type 2. EMBO Mol Med 8(8):967–986PubMedPubMedCentralCrossRef
86.
go back to reference Bottger P, Glerup S, Gesslein B, Illarionova NB, Isaksen TJ, Heuck A et al (2016) Glutamate-system defects behind psychiatric manifestations in a familial hemiplegic migraine type 2 disease-mutation mouse model. Sci Rep 6:22047PubMedPubMedCentralCrossRef Bottger P, Glerup S, Gesslein B, Illarionova NB, Isaksen TJ, Heuck A et al (2016) Glutamate-system defects behind psychiatric manifestations in a familial hemiplegic migraine type 2 disease-mutation mouse model. Sci Rep 6:22047PubMedPubMedCentralCrossRef
88.
go back to reference Isaksen TJ, Lykke-Hartmann K (2016) Insights into the pathology of the alpha2-Na(+)/K(+)-ATPase in neurological disorders; lessons from animal models. Front Physiol 7:161PubMedPubMedCentralCrossRef Isaksen TJ, Lykke-Hartmann K (2016) Insights into the pathology of the alpha2-Na(+)/K(+)-ATPase in neurological disorders; lessons from animal models. Front Physiol 7:161PubMedPubMedCentralCrossRef
89.
90.
go back to reference Dichgans M, Freilinger T, Eckstein G, Babini E, Lorenz-Depiereux B, Biskup S et al (2005) Mutation in the neuronal voltage-gated sodium channel SCN1A in familial hemiplegic migraine. Lancet 366(9483):371–377PubMedCrossRef Dichgans M, Freilinger T, Eckstein G, Babini E, Lorenz-Depiereux B, Biskup S et al (2005) Mutation in the neuronal voltage-gated sodium channel SCN1A in familial hemiplegic migraine. Lancet 366(9483):371–377PubMedCrossRef
91.
go back to reference Catterall WA (2000) From ionic currents to molecular mechanisms: the structure and function of voltage-gated sodium channels. Neuron 26(1):13–25PubMedCrossRef Catterall WA (2000) From ionic currents to molecular mechanisms: the structure and function of voltage-gated sodium channels. Neuron 26(1):13–25PubMedCrossRef
92.
go back to reference Meng H, Xu HQ, Yu L, Lin GW, He N, Su T et al (2015) The SCN1A mutation database: updating information and analysis of the relationships among genotype, functional alteration, and phenotype. Hum Mutat 36(6):573–580PubMedCrossRef Meng H, Xu HQ, Yu L, Lin GW, He N, Su T et al (2015) The SCN1A mutation database: updating information and analysis of the relationships among genotype, functional alteration, and phenotype. Hum Mutat 36(6):573–580PubMedCrossRef
93.
go back to reference Sutherland HG, Griffiths LR (2017) Genetics of migraine: insights into the molecular basis of migraine disorders. Headache 57(4):537–569PubMedCrossRef Sutherland HG, Griffiths LR (2017) Genetics of migraine: insights into the molecular basis of migraine disorders. Headache 57(4):537–569PubMedCrossRef
94.
go back to reference Zhang Y, Chen N, Zhou M, Guo J, Guo J, He L (2017) A novel SCN1A mutation identified in a Chinese family with familial hemiplegic migraine: a case report. Cephalalgia 37(13):1294–1298PubMedCrossRef Zhang Y, Chen N, Zhou M, Guo J, Guo J, He L (2017) A novel SCN1A mutation identified in a Chinese family with familial hemiplegic migraine: a case report. Cephalalgia 37(13):1294–1298PubMedCrossRef
95.
go back to reference Mantegazza M, Cestele S (2018) Pathophysiological mechanisms of migraine and epilepsy: similarities and differences. Neurosci Lett 667:92–102PubMedCrossRef Mantegazza M, Cestele S (2018) Pathophysiological mechanisms of migraine and epilepsy: similarities and differences. Neurosci Lett 667:92–102PubMedCrossRef
96.
go back to reference Le Fort D, Safran AB, Picard F, Bouchardy I, Morris MA (2004) Elicited repetitive daily blindness: a new familial disorder related to migraine and epilepsy. Neurology 63(2):348–350PubMedCrossRef Le Fort D, Safran AB, Picard F, Bouchardy I, Morris MA (2004) Elicited repetitive daily blindness: a new familial disorder related to migraine and epilepsy. Neurology 63(2):348–350PubMedCrossRef
97.
go back to reference Castro MJ, Stam AH, Lemos C, de Vries B, Vanmolkot KR, Barros J et al (2009) First mutation in the voltage-gated Nav1.1 subunit gene SCN1A with co-occurring familial hemiplegic migraine and epilepsy. Cephalalgia 29(3):308–313PubMedCrossRef Castro MJ, Stam AH, Lemos C, de Vries B, Vanmolkot KR, Barros J et al (2009) First mutation in the voltage-gated Nav1.1 subunit gene SCN1A with co-occurring familial hemiplegic migraine and epilepsy. Cephalalgia 29(3):308–313PubMedCrossRef
98.
go back to reference Vanmolkot KR, Babini E, de Vries B, Stam AH, Freilinger T, Terwindt GM et al (2007) The novel p.L1649Q mutation in the SCN1A epilepsy gene is associated with familial hemiplegic migraine: genetic and functional studies. Mutation in brief #957. Online. Hum Mutat 28(5):522PubMedCrossRef Vanmolkot KR, Babini E, de Vries B, Stam AH, Freilinger T, Terwindt GM et al (2007) The novel p.L1649Q mutation in the SCN1A epilepsy gene is associated with familial hemiplegic migraine: genetic and functional studies. Mutation in brief #957. Online. Hum Mutat 28(5):522PubMedCrossRef
99.
go back to reference Catterall WA (2018) Dravet syndrome: a Sodium Channel Interneuronopathy. Curr Opin Physiol 2:42–50PubMedCrossRef Catterall WA (2018) Dravet syndrome: a Sodium Channel Interneuronopathy. Curr Opin Physiol 2:42–50PubMedCrossRef
101.
go back to reference Marini C, Scheffer IE, Nabbout R, Suls A, De Jonghe P, Zara F et al (2011) The genetics of Dravet syndrome. Epilepsia 52(Suppl 2):24–29PubMedCrossRef Marini C, Scheffer IE, Nabbout R, Suls A, De Jonghe P, Zara F et al (2011) The genetics of Dravet syndrome. Epilepsia 52(Suppl 2):24–29PubMedCrossRef
102.
go back to reference Ogiwara I, Miyamoto H, Morita N, Atapour N, Mazaki E, Inoue I et al (2007) Nav1.1 localizes to axons of parvalbumin-positive inhibitory interneurons: a circuit basis for epileptic seizures in mice carrying an Scn1a gene mutation. J Neurosci 27(22):5903–5914PubMedCrossRefPubMedCentral Ogiwara I, Miyamoto H, Morita N, Atapour N, Mazaki E, Inoue I et al (2007) Nav1.1 localizes to axons of parvalbumin-positive inhibitory interneurons: a circuit basis for epileptic seizures in mice carrying an Scn1a gene mutation. J Neurosci 27(22):5903–5914PubMedCrossRefPubMedCentral
103.
go back to reference Yu FH, Mantegazza M, Westenbroek RE, Robbins CA, Kalume F, Burton KA et al (2006) Reduced sodium current in GABAergic interneurons in a mouse model of severe myoclonic epilepsy in infancy. Nat Neurosci 9(9):1142–1149PubMedCrossRef Yu FH, Mantegazza M, Westenbroek RE, Robbins CA, Kalume F, Burton KA et al (2006) Reduced sodium current in GABAergic interneurons in a mouse model of severe myoclonic epilepsy in infancy. Nat Neurosci 9(9):1142–1149PubMedCrossRef
104.
go back to reference Cestele S, Scalmani P, Rusconi R, Terragni B, Franceschetti S, Mantegazza M (2008) Self-limited hyperexcitability: functional effect of a familial hemiplegic migraine mutation of the Nav1.1 (SCN1A) Na+ channel. J Neurosci 28(29):7273–7283PubMedPubMedCentralCrossRef Cestele S, Scalmani P, Rusconi R, Terragni B, Franceschetti S, Mantegazza M (2008) Self-limited hyperexcitability: functional effect of a familial hemiplegic migraine mutation of the Nav1.1 (SCN1A) Na+ channel. J Neurosci 28(29):7273–7283PubMedPubMedCentralCrossRef
105.
go back to reference Cestele S, Labate A, Rusconi R, Tarantino P, Mumoli L, Franceschetti S et al (2013) Divergent effects of the T1174S SCN1A mutation associated with seizures and hemiplegic migraine. Epilepsia 54(5):927–935PubMedCrossRef Cestele S, Labate A, Rusconi R, Tarantino P, Mumoli L, Franceschetti S et al (2013) Divergent effects of the T1174S SCN1A mutation associated with seizures and hemiplegic migraine. Epilepsia 54(5):927–935PubMedCrossRef
106.
go back to reference Fan C, Wolking S, Lehmann-Horn F, Hedrich UB, Freilinger T, Lerche H et al (2016) Early-onset familial hemiplegic migraine due to a novel SCN1A mutation. Cephalalgia 36(13):1238-1247PubMedPubMedCentralCrossRef Fan C, Wolking S, Lehmann-Horn F, Hedrich UB, Freilinger T, Lerche H et al (2016) Early-onset familial hemiplegic migraine due to a novel SCN1A mutation. Cephalalgia 36(13):1238-1247PubMedPubMedCentralCrossRef
107.
go back to reference Bertelli S, Barbieri R, Pusch M, Gavazzo P (2018) Gain of function of sporadic/familial hemiplegic migraine-causing SCN1A mutations: use of an optimized cDNA. Cephalalgia 39(4):477-488PubMedCrossRef Bertelli S, Barbieri R, Pusch M, Gavazzo P (2018) Gain of function of sporadic/familial hemiplegic migraine-causing SCN1A mutations: use of an optimized cDNA. Cephalalgia 39(4):477-488PubMedCrossRef
108.
go back to reference Pellacani S, Sicca F, Di Lorenzo C, Grieco GS, Valvo G, Cereda C et al (2016) The revolution in migraine genetics: from aching channels disorders to a next-generation medicine. Front Cell Neurosci 10:156PubMedPubMedCentralCrossRef Pellacani S, Sicca F, Di Lorenzo C, Grieco GS, Valvo G, Cereda C et al (2016) The revolution in migraine genetics: from aching channels disorders to a next-generation medicine. Front Cell Neurosci 10:156PubMedPubMedCentralCrossRef
109.
go back to reference Kahlig KM, Rhodes TH, Pusch M, Freilinger T, Pereira-Monteiro JM, Ferrari MD et al (2008) Divergent sodium channel defects in familial hemiplegic migraine. Proc Natl Acad Sci U S A 105(28):9799–9804PubMedPubMedCentralCrossRef Kahlig KM, Rhodes TH, Pusch M, Freilinger T, Pereira-Monteiro JM, Ferrari MD et al (2008) Divergent sodium channel defects in familial hemiplegic migraine. Proc Natl Acad Sci U S A 105(28):9799–9804PubMedPubMedCentralCrossRef
110.
go back to reference Cestele S, Schiavon E, Rusconi R, Franceschetti S, Mantegazza M (2013) Nonfunctional NaV1.1 familial hemiplegic migraine mutant transformed into gain of function by partial rescue of folding defects. Proc Natl Acad Sci U S A 110(43):17546–17551PubMedPubMedCentralCrossRef Cestele S, Schiavon E, Rusconi R, Franceschetti S, Mantegazza M (2013) Nonfunctional NaV1.1 familial hemiplegic migraine mutant transformed into gain of function by partial rescue of folding defects. Proc Natl Acad Sci U S A 110(43):17546–17551PubMedPubMedCentralCrossRef
111.
go back to reference Dhifallah S, Lancaster E, Merrill S, Leroudier N, Mantegazza M, Cestele S (2018) Gain of function for the SCN1A/hNav1.1-L1670W mutation responsible for familial hemiplegic migraine. Front Mol Neurosci 11:232PubMedPubMedCentralCrossRef Dhifallah S, Lancaster E, Merrill S, Leroudier N, Mantegazza M, Cestele S (2018) Gain of function for the SCN1A/hNav1.1-L1670W mutation responsible for familial hemiplegic migraine. Front Mol Neurosci 11:232PubMedPubMedCentralCrossRef
112.
go back to reference de Vries B, Freilinger T, Vanmolkot KR, Koenderink JB, Stam AH, Terwindt GM et al (2007) Systematic analysis of three FHM genes in 39 sporadic patients with hemiplegic migraine. Neurology 69(23):2170–2176PubMedCrossRef de Vries B, Freilinger T, Vanmolkot KR, Koenderink JB, Stam AH, Terwindt GM et al (2007) Systematic analysis of three FHM genes in 39 sporadic patients with hemiplegic migraine. Neurology 69(23):2170–2176PubMedCrossRef
113.
go back to reference Silveira-Moriyama L, Kovac S, Kurian MA, Houlden H, Lees AJ, Walker MC et al (2018) Phenotypes, genotypes, and the management of paroxysmal movement disorders. Dev Med Child Neurol 60(6):559–565CrossRefPubMed Silveira-Moriyama L, Kovac S, Kurian MA, Houlden H, Lees AJ, Walker MC et al (2018) Phenotypes, genotypes, and the management of paroxysmal movement disorders. Dev Med Child Neurol 60(6):559–565CrossRefPubMed
114.
go back to reference Lee HY, Xu Y, Huang Y, Ahn AH, Auburger GW, Pandolfo M et al (2004) The gene for paroxysmal non-kinesigenic dyskinesia encodes an enzyme in a stress response pathway. Hum Mol Genet 13(24):3161–3170PubMedCrossRef Lee HY, Xu Y, Huang Y, Ahn AH, Auburger GW, Pandolfo M et al (2004) The gene for paroxysmal non-kinesigenic dyskinesia encodes an enzyme in a stress response pathway. Hum Mol Genet 13(24):3161–3170PubMedCrossRef
115.
go back to reference Rainier S, Thomas D, Tokarz D, Ming L, Bui M, Plein E et al (2004) Myofibrillogenesis regulator 1 gene mutations cause paroxysmal dystonic choreoathetosis. Arch Neurol 61(7):1025–1029CrossRefPubMed Rainier S, Thomas D, Tokarz D, Ming L, Bui M, Plein E et al (2004) Myofibrillogenesis regulator 1 gene mutations cause paroxysmal dystonic choreoathetosis. Arch Neurol 61(7):1025–1029CrossRefPubMed
116.
go back to reference Chen W-J, Lin Y, Xiong Z-Q, Wei W, Ni W, Tan G-H et al (2011) Exome sequencing identifies truncating mutations in PRRT2 that cause paroxysmal kinesigenic dyskinesia. Nat Genet 43:1252CrossRefPubMed Chen W-J, Lin Y, Xiong Z-Q, Wei W, Ni W, Tan G-H et al (2011) Exome sequencing identifies truncating mutations in PRRT2 that cause paroxysmal kinesigenic dyskinesia. Nat Genet 43:1252CrossRefPubMed
117.
go back to reference Wang J-L, Cao L, Li X-H, Hu Z-M, Li J-D, Zhang J-G et al (2011) Identification of PRRT2 as the causative gene of paroxysmal kinesigenic dyskinesias. Brain 134(12):3493–3501PubMedCrossRef Wang J-L, Cao L, Li X-H, Hu Z-M, Li J-D, Zhang J-G et al (2011) Identification of PRRT2 as the causative gene of paroxysmal kinesigenic dyskinesias. Brain 134(12):3493–3501PubMedCrossRef
118.
go back to reference Gardiner AR, Jaffer F, Dale RC, Labrum R, Erro R, Meyer E et al (2015) The clinical and genetic heterogeneity of paroxysmal dyskinesias. Brain 138(Pt 12):3567–3580PubMedPubMedCentralCrossRef Gardiner AR, Jaffer F, Dale RC, Labrum R, Erro R, Meyer E et al (2015) The clinical and genetic heterogeneity of paroxysmal dyskinesias. Brain 138(Pt 12):3567–3580PubMedPubMedCentralCrossRef
119.
go back to reference Heron SE, Grinton BE, Kivity S, Afawi Z, Zuberi SM, Hughes JN et al (2012) PRRT2 mutations cause benign familial infantile epilepsy and infantile convulsions with choreoathetosis syndrome. Am J Hum Genet 90(1):152–160PubMedPubMedCentralCrossRef Heron SE, Grinton BE, Kivity S, Afawi Z, Zuberi SM, Hughes JN et al (2012) PRRT2 mutations cause benign familial infantile epilepsy and infantile convulsions with choreoathetosis syndrome. Am J Hum Genet 90(1):152–160PubMedPubMedCentralCrossRef
120.
go back to reference Ono S, Yoshiura K, Kinoshita A, Kikuchi T, Nakane Y, Kato N et al (2012) Mutations in PRRT2 responsible for paroxysmal kinesigenic dyskinesias also cause benign familial infantile convulsions. J Hum Genet 57(5):338–341PubMedCrossRef Ono S, Yoshiura K, Kinoshita A, Kikuchi T, Nakane Y, Kato N et al (2012) Mutations in PRRT2 responsible for paroxysmal kinesigenic dyskinesias also cause benign familial infantile convulsions. J Hum Genet 57(5):338–341PubMedCrossRef
121.
go back to reference Riant F, Roze E, Barbance C, Meneret A, Guyant-Marechal L, Lucas C et al (2012) PRRT2 mutations cause hemiplegic migraine. Neurology 79(21):2122–2124PubMedCrossRef Riant F, Roze E, Barbance C, Meneret A, Guyant-Marechal L, Lucas C et al (2012) PRRT2 mutations cause hemiplegic migraine. Neurology 79(21):2122–2124PubMedCrossRef
122.
go back to reference Marini C, Conti V, Mei D, Battaglia D, Lettori D, Losito E et al (2012) PRRT2 mutations in familial infantile seizures, paroxysmal dyskinesia, and hemiplegic migraine. Neurology 79(21):2109–2114PubMedPubMedCentralCrossRef Marini C, Conti V, Mei D, Battaglia D, Lettori D, Losito E et al (2012) PRRT2 mutations in familial infantile seizures, paroxysmal dyskinesia, and hemiplegic migraine. Neurology 79(21):2109–2114PubMedPubMedCentralCrossRef
123.
go back to reference Dale RC, Gardiner A, Antony J, Houlden H (2012) Familial PRRT2 mutation with heterogeneous paroxysmal disorders including paroxysmal torticollis and hemiplegic migraine. Dev Med Child Neurol 54(10):958–960PubMedCrossRef Dale RC, Gardiner A, Antony J, Houlden H (2012) Familial PRRT2 mutation with heterogeneous paroxysmal disorders including paroxysmal torticollis and hemiplegic migraine. Dev Med Child Neurol 54(10):958–960PubMedCrossRef
124.
go back to reference Ebrahimi-Fakhari D, Saffari A, Westenberger A, Klein C (2015) The evolving spectrum of PRRT2-associated paroxysmal diseases. Brain 138(Pt 12):3476–3495PubMedCrossRef Ebrahimi-Fakhari D, Saffari A, Westenberger A, Klein C (2015) The evolving spectrum of PRRT2-associated paroxysmal diseases. Brain 138(Pt 12):3476–3495PubMedCrossRef
125.
go back to reference Pelzer N, de Vries B, Kamphorst JT, Vijfhuizen LS, Ferrari MD, Haan J et al (2014) PRRT2 and hemiplegic migraine: a complex association. Neurology 83(3):288–290PubMedCrossRef Pelzer N, de Vries B, Kamphorst JT, Vijfhuizen LS, Ferrari MD, Haan J et al (2014) PRRT2 and hemiplegic migraine: a complex association. Neurology 83(3):288–290PubMedCrossRef
126.
go back to reference Lee HY, Huang Y, Bruneau N, Roll P, Roberson ED, Hermann M et al (2012) Mutations in the gene PRRT2 cause paroxysmal kinesigenic dyskinesia with infantile convulsions. Cell Rep 1(1):2–12PubMedCrossRef Lee HY, Huang Y, Bruneau N, Roll P, Roberson ED, Hermann M et al (2012) Mutations in the gene PRRT2 cause paroxysmal kinesigenic dyskinesia with infantile convulsions. Cell Rep 1(1):2–12PubMedCrossRef
127.
go back to reference Valente P, Castroflorio E, Rossi P, Fadda M, Sterlini B, Cervigni RI et al (2016) PRRT2 is a key component of the ca (2+)-dependent neurotransmitter release machinery. Cell Rep 15(1):117–131PubMedPubMedCentralCrossRef Valente P, Castroflorio E, Rossi P, Fadda M, Sterlini B, Cervigni RI et al (2016) PRRT2 is a key component of the ca (2+)-dependent neurotransmitter release machinery. Cell Rep 15(1):117–131PubMedPubMedCentralCrossRef
128.
go back to reference Fruscione F, Valente P, Sterlini B, Romei A, Baldassari S, Fadda M et al (2018) PRRT2 controls neuronal excitability by negatively modulating Na+ channel 1.2/1.6 activity. Brain 141(4):1000–1016PubMedPubMedCentralCrossRef Fruscione F, Valente P, Sterlini B, Romei A, Baldassari S, Fadda M et al (2018) PRRT2 controls neuronal excitability by negatively modulating Na+ channel 1.2/1.6 activity. Brain 141(4):1000–1016PubMedPubMedCentralCrossRef
129.
go back to reference Condliffe SB, Corradini I, Pozzi D, Verderio C, Matteoli M (2010) Endogenous SNAP-25 regulates native voltage-gated calcium channels in glutamatergic neurons. J Biol Chem 285(32):24968–24976PubMedPubMedCentralCrossRef Condliffe SB, Corradini I, Pozzi D, Verderio C, Matteoli M (2010) Endogenous SNAP-25 regulates native voltage-gated calcium channels in glutamatergic neurons. J Biol Chem 285(32):24968–24976PubMedPubMedCentralCrossRef
130.
go back to reference Weller CM, Leen WG, Neville BG, Duncan JS, de Vries B, Geilenkirchen MA et al (2015) A novel SLC2A1 mutation linking hemiplegic migraine with alternating hemiplegia of childhood. Cephalalgia 35(1):10–15PubMedCrossRef Weller CM, Leen WG, Neville BG, Duncan JS, de Vries B, Geilenkirchen MA et al (2015) A novel SLC2A1 mutation linking hemiplegic migraine with alternating hemiplegia of childhood. Cephalalgia 35(1):10–15PubMedCrossRef
131.
go back to reference Mohammad SS, Coman D, Calvert S (2014) Glucose transporter 1 deficiency syndrome and hemiplegic migraines as a dominant presenting clinical feature. J Paediatr Child Health 50(12):1025–1026PubMed Mohammad SS, Coman D, Calvert S (2014) Glucose transporter 1 deficiency syndrome and hemiplegic migraines as a dominant presenting clinical feature. J Paediatr Child Health 50(12):1025–1026PubMed
132.
go back to reference Jen JC, Wan J, Palos TP, Howard BD, Baloh RW (2005) Mutation in the glutamate transporter EAAT1 causes episodic ataxia, hemiplegia, and seizures. Neurology 65(4):529–534PubMedCrossRef Jen JC, Wan J, Palos TP, Howard BD, Baloh RW (2005) Mutation in the glutamate transporter EAAT1 causes episodic ataxia, hemiplegia, and seizures. Neurology 65(4):529–534PubMedCrossRef
133.
go back to reference Kovermann P, Hessel M, Kortzak D, Jen JC, Koch J, Fahlke C et al (2017) Impaired K(+) binding to glial glutamate transporter EAAT1 in migraine. Sci Rep 7(1):13913PubMedPubMedCentralCrossRef Kovermann P, Hessel M, Kortzak D, Jen JC, Koch J, Fahlke C et al (2017) Impaired K(+) binding to glial glutamate transporter EAAT1 in migraine. Sci Rep 7(1):13913PubMedPubMedCentralCrossRef
134.
go back to reference Suzuki M, Van Paesschen W, Stalmans I, Horita S, Yamada H, Bergmans BA et al (2010) Defective membrane expression of the Na(+)-HCO (3)(−) cotransporter NBCe1 is associated with familial migraine. Proc Natl Acad Sci U S A 107(36):15963–15968PubMedPubMedCentralCrossRef Suzuki M, Van Paesschen W, Stalmans I, Horita S, Yamada H, Bergmans BA et al (2010) Defective membrane expression of the Na(+)-HCO (3)(−) cotransporter NBCe1 is associated with familial migraine. Proc Natl Acad Sci U S A 107(36):15963–15968PubMedPubMedCentralCrossRef
135.
go back to reference Lafreniere RG, Cader MZ, Poulin JF, Andres-Enguix I, Simoneau M, Gupta N et al (2010) A dominant-negative mutation in the TRESK potassium channel is linked to familial migraine with aura. Nat Med 16(10):1157–1160PubMedCrossRef Lafreniere RG, Cader MZ, Poulin JF, Andres-Enguix I, Simoneau M, Gupta N et al (2010) A dominant-negative mutation in the TRESK potassium channel is linked to familial migraine with aura. Nat Med 16(10):1157–1160PubMedCrossRef
136.
go back to reference Kang D, Kim D (2006) TREK-2 (K2P10.1) and TRESK (K2P18.1) are major background K+ channels in dorsal root ganglion neurons. Am J Phys Cell Physiol 291(1):C138–C146CrossRef Kang D, Kim D (2006) TREK-2 (K2P10.1) and TRESK (K2P18.1) are major background K+ channels in dorsal root ganglion neurons. Am J Phys Cell Physiol 291(1):C138–C146CrossRef
137.
go back to reference Enyedi P, Czirjak G (2015) Properties, regulation, pharmacology, and functions of the K (2) p channel, TRESK. Pflugers Arch 467(5):945–958PubMedCrossRef Enyedi P, Czirjak G (2015) Properties, regulation, pharmacology, and functions of the K (2) p channel, TRESK. Pflugers Arch 467(5):945–958PubMedCrossRef
138.
go back to reference Dobler T, Springauf A, Tovornik S, Weber M, Schmitt A, Sedlmeier R et al (2007) TRESK two-pore-domain K+ channels constitute a significant component of background potassium currents in murine dorsal root ganglion neurones. J Physiol 585(Pt 3):867–879PubMedPubMedCentralCrossRef Dobler T, Springauf A, Tovornik S, Weber M, Schmitt A, Sedlmeier R et al (2007) TRESK two-pore-domain K+ channels constitute a significant component of background potassium currents in murine dorsal root ganglion neurones. J Physiol 585(Pt 3):867–879PubMedPubMedCentralCrossRef
139.
go back to reference Chae YJ, Zhang J, Au P, Sabbadini M, Xie GX, Yost CS (2010) Discrete change in volatile anesthetic sensitivity in mice with inactivated tandem pore potassium ion channel TRESK. Anesthesiology 113(6):1326–1337PubMedCrossRef Chae YJ, Zhang J, Au P, Sabbadini M, Xie GX, Yost CS (2010) Discrete change in volatile anesthetic sensitivity in mice with inactivated tandem pore potassium ion channel TRESK. Anesthesiology 113(6):1326–1337PubMedCrossRef
140.
go back to reference Liu P, Xiao Z, Ren F, Guo Z, Chen Z, Zhao H et al (2013) Functional analysis of a migraine-associated TRESK K+ channel mutation. J Neurosci 33(31):12810–12824PubMedPubMedCentralCrossRef Liu P, Xiao Z, Ren F, Guo Z, Chen Z, Zhao H et al (2013) Functional analysis of a migraine-associated TRESK K+ channel mutation. J Neurosci 33(31):12810–12824PubMedPubMedCentralCrossRef
141.
go back to reference Guo Z, Liu P, Ren F, Cao YQ (2014) Nonmigraine-associated TRESK K+ channel variant C110R does not increase the excitability of trigeminal ganglion neurons. J Neurophysiol 112(3):568–579PubMedPubMedCentralCrossRef Guo Z, Liu P, Ren F, Cao YQ (2014) Nonmigraine-associated TRESK K+ channel variant C110R does not increase the excitability of trigeminal ganglion neurons. J Neurophysiol 112(3):568–579PubMedPubMedCentralCrossRef
142.
go back to reference Andres-Enguix I, Shang L, Stansfeld PJ, Morahan JM, Sansom MS, Lafreniere RG et al (2012) Functional analysis of missense variants in the TRESK (KCNK18) K channel. Sci Rep 2:237PubMedPubMedCentralCrossRef Andres-Enguix I, Shang L, Stansfeld PJ, Morahan JM, Sansom MS, Lafreniere RG et al (2012) Functional analysis of missense variants in the TRESK (KCNK18) K channel. Sci Rep 2:237PubMedPubMedCentralCrossRef
143.
go back to reference Royal P, Andres-Bilbe A, Avalos Prado P, Verkest C, Wdziekonski B, Schaub S et al (2019) Migraine-associated TRESK mutations increase neuronal excitability through alternative translation initiation and inhibition of TREK. Neuron 101(2):232–45 e6PubMedCrossRef Royal P, Andres-Bilbe A, Avalos Prado P, Verkest C, Wdziekonski B, Schaub S et al (2019) Migraine-associated TRESK mutations increase neuronal excitability through alternative translation initiation and inhibition of TREK. Neuron 101(2):232–45 e6PubMedCrossRef
144.
go back to reference Brennan KC, Bates EA, Shapiro RE, Zyuzin J, Hallows WC, Huang Y et al (2013) Casein kinase idelta mutations in familial migraine and advanced sleep phase. Sci Transl Med 5(183):183ra56, 1–183ra56,11CrossRef Brennan KC, Bates EA, Shapiro RE, Zyuzin J, Hallows WC, Huang Y et al (2013) Casein kinase idelta mutations in familial migraine and advanced sleep phase. Sci Transl Med 5(183):183ra56, 1–183ra56,11CrossRef
145.
go back to reference Xu Y, Padiath QS, Shapiro RE, Jones CR, Wu SC, Saigoh N et al (2005) Functional consequences of a CKIdelta mutation causing familial advanced sleep phase syndrome. Nature 434(7033):640–644PubMedCrossRef Xu Y, Padiath QS, Shapiro RE, Jones CR, Wu SC, Saigoh N et al (2005) Functional consequences of a CKIdelta mutation causing familial advanced sleep phase syndrome. Nature 434(7033):640–644PubMedCrossRef
146.
go back to reference Xu Y, Toh KL, Jones CR, Shin JY, Fu YH, Ptacek LJ (2007) Modeling of a human circadian mutation yields insights into clock regulation by PER2. Cell 128(1):59–70PubMedPubMedCentralCrossRef Xu Y, Toh KL, Jones CR, Shin JY, Fu YH, Ptacek LJ (2007) Modeling of a human circadian mutation yields insights into clock regulation by PER2. Cell 128(1):59–70PubMedPubMedCentralCrossRef
149.
go back to reference Maniyar FH, Sprenger T, Monteith T, Schankin C, Goadsby PJ (2014) Brain activations in the premonitory phase of nitroglycerin-triggered migraine attacks. Brain 137(Pt 1):232–241PubMedCrossRef Maniyar FH, Sprenger T, Monteith T, Schankin C, Goadsby PJ (2014) Brain activations in the premonitory phase of nitroglycerin-triggered migraine attacks. Brain 137(Pt 1):232–241PubMedCrossRef
150.
go back to reference Williams LB, Javed A, Sabri A, Morgan DJ, Huff CD, Grigg JR et al (2019) ALPK1 missense pathogenic variant in five families leads to ROSAH syndrome, an ocular multisystem autosomal dominant disorder. Genet Med. https://doi.org/10.1038/s41436-019-0476-3. [Epub ahead of print] Williams LB, Javed A, Sabri A, Morgan DJ, Huff CD, Grigg JR et al (2019) ALPK1 missense pathogenic variant in five families leads to ROSAH syndrome, an ocular multisystem autosomal dominant disorder. Genet Med. https://​doi.​org/​10.​1038/​s41436-019-0476-3. [Epub ahead of print]
151.
go back to reference Dichgans M, Mayer M, Uttner I, Bruning R, Muller-Hocker J, Rungger G et al (1998) The phenotypic spectrum of CADASIL: clinical findings in 102 cases. Ann Neurol 44(5):731–739PubMedCrossRef Dichgans M, Mayer M, Uttner I, Bruning R, Muller-Hocker J, Rungger G et al (1998) The phenotypic spectrum of CADASIL: clinical findings in 102 cases. Ann Neurol 44(5):731–739PubMedCrossRef
152.
go back to reference Guey S, Mawet J, Herve D, Duering M, Godin O, Jouvent E et al (2016) Prevalence and characteristics of migraine in CADASIL. Cephalalgia 36(11):1038–1047PubMedCrossRef Guey S, Mawet J, Herve D, Duering M, Godin O, Jouvent E et al (2016) Prevalence and characteristics of migraine in CADASIL. Cephalalgia 36(11):1038–1047PubMedCrossRef
154.
go back to reference Liem MK, Oberstein SA, van der Grond J, Ferrari MD, Haan JCADASIL (2010) Migraine: a narrative review. Cephalalgia 30(11):1284–1289PubMedCrossRef Liem MK, Oberstein SA, van der Grond J, Ferrari MD, Haan JCADASIL (2010) Migraine: a narrative review. Cephalalgia 30(11):1284–1289PubMedCrossRef
155.
go back to reference Rossi G, Shambhu S (2018) Hemiplegic migraine as the initial presentation of biopsy positive cerebral autosomal dominant Arteriopathy with subcortical infarcts and leukoencephalopathy. Cureus 10(5):e2631PubMedPubMedCentral Rossi G, Shambhu S (2018) Hemiplegic migraine as the initial presentation of biopsy positive cerebral autosomal dominant Arteriopathy with subcortical infarcts and leukoencephalopathy. Cureus 10(5):e2631PubMedPubMedCentral
156.
go back to reference Rice GI, Rodero MP, Crow YJ (2015) Human disease phenotypes associated with mutations in TREX1. J Clin Immunol 35(3):235–243PubMedCrossRef Rice GI, Rodero MP, Crow YJ (2015) Human disease phenotypes associated with mutations in TREX1. J Clin Immunol 35(3):235–243PubMedCrossRef
157.
go back to reference Stam AH, Kothari PH, Shaikh A, Gschwendter A, Jen JC, Hodgkinson S et al (2016) Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations. Brain 139(11):2909-2922 Stam AH, Kothari PH, Shaikh A, Gschwendter A, Jen JC, Hodgkinson S et al (2016) Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations. Brain 139(11):2909-2922
158.
go back to reference Stam AH, Haan J, van den Maagdenberg AM, Ferrari MD, Terwindt GM (2009) Migraine and genetic and acquired vasculopathies. Cephalalgia 29(9):1006–1017PubMedCrossRef Stam AH, Haan J, van den Maagdenberg AM, Ferrari MD, Terwindt GM (2009) Migraine and genetic and acquired vasculopathies. Cephalalgia 29(9):1006–1017PubMedCrossRef
159.
go back to reference Breedveld G, de Coo IF, Lequin MH, Arts WF, Heutink P, Gould DB et al (2006) Novel mutations in three families confirm a major role of COL4A1 in hereditary porencephaly. J Med Genet 43(6):490–495PubMedCrossRef Breedveld G, de Coo IF, Lequin MH, Arts WF, Heutink P, Gould DB et al (2006) Novel mutations in three families confirm a major role of COL4A1 in hereditary porencephaly. J Med Genet 43(6):490–495PubMedCrossRef
160.
go back to reference Sondergaard CB, Nielsen JE, Hansen CK, Christensen H (2017) Hereditary cerebral small vessel disease and stroke. Clin Neurol Neurosurg 155:45–57PubMedCrossRef Sondergaard CB, Nielsen JE, Hansen CK, Christensen H (2017) Hereditary cerebral small vessel disease and stroke. Clin Neurol Neurosurg 155:45–57PubMedCrossRef
161.
go back to reference Gormley P, Winsvold BS, Nyholt DR, Kallela M, Chasman DI, Palotie A (2016) Migraine genetics: from genome-wide association studies to translational insights. Genome Med 8(1):86PubMedPubMedCentralCrossRef Gormley P, Winsvold BS, Nyholt DR, Kallela M, Chasman DI, Palotie A (2016) Migraine genetics: from genome-wide association studies to translational insights. Genome Med 8(1):86PubMedPubMedCentralCrossRef
164.
go back to reference Hu Y, Jiang H, Wang Q, Xie Z, Pan S (2013) Identification of a novel nonsense mutation p.Tyr1957Ter of CACNA1A in a Chinese family with episodic ataxia 2. PLoS One 8(2):e56362PubMedPubMedCentralCrossRef Hu Y, Jiang H, Wang Q, Xie Z, Pan S (2013) Identification of a novel nonsense mutation p.Tyr1957Ter of CACNA1A in a Chinese family with episodic ataxia 2. PLoS One 8(2):e56362PubMedPubMedCentralCrossRef
165.
go back to reference Oh SK, Baek JI, Weigand KM, Venselaar H, Swarts HG, Park SH et al (2015) A missense variant of the ATP1A2 gene is associated with a novel phenotype of progressive sensorineural hearing loss associated with migraine. Eur J Hum Genet 23(5):639–645PubMedCrossRef Oh SK, Baek JI, Weigand KM, Venselaar H, Swarts HG, Park SH et al (2015) A missense variant of the ATP1A2 gene is associated with a novel phenotype of progressive sensorineural hearing loss associated with migraine. Eur J Hum Genet 23(5):639–645PubMedCrossRef
166.
go back to reference Maksemous N, Roy B, Smith RA, Griffiths LR (2016) Next-generation sequencing identifies novel CACNA1A gene mutations in episodic ataxia type 2. Mol Genet Genomic Med 4(2):211–222PubMedPubMedCentralCrossRef Maksemous N, Roy B, Smith RA, Griffiths LR (2016) Next-generation sequencing identifies novel CACNA1A gene mutations in episodic ataxia type 2. Mol Genet Genomic Med 4(2):211–222PubMedPubMedCentralCrossRef
167.
go back to reference Maksemous N, Smith RA, Haupt LM, Griffiths LR (2016) Targeted next generation sequencing identifies novel NOTCH3 gene mutations in CADASIL diagnostics patients. Hum Genomics 10(1):38PubMedPubMedCentralCrossRef Maksemous N, Smith RA, Haupt LM, Griffiths LR (2016) Targeted next generation sequencing identifies novel NOTCH3 gene mutations in CADASIL diagnostics patients. Hum Genomics 10(1):38PubMedPubMedCentralCrossRef
168.
go back to reference Klassen T, Davis C, Goldman A, Burgess D, Chen T, Wheeler D et al (2011) Exome sequencing of ion channel genes reveals complex profiles confounding personal risk assessment in epilepsy. Cell 145(7):1036–1048PubMedPubMedCentralCrossRef Klassen T, Davis C, Goldman A, Burgess D, Chen T, Wheeler D et al (2011) Exome sequencing of ion channel genes reveals complex profiles confounding personal risk assessment in epilepsy. Cell 145(7):1036–1048PubMedPubMedCentralCrossRef
169.
go back to reference Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T et al (2016) Analysis of protein-coding genetic variation in 60,706 humans. Nature 536(7616):285–291PubMedPubMedCentralCrossRef Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T et al (2016) Analysis of protein-coding genetic variation in 60,706 humans. Nature 536(7616):285–291PubMedPubMedCentralCrossRef
170.
go back to reference Landrum MJ, Lee JM, Benson M, Brown G, Chao C, Chitipiralla S et al (2016) ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res 44(D1):D862–D868PubMedCrossRef Landrum MJ, Lee JM, Benson M, Brown G, Chao C, Chitipiralla S et al (2016) ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res 44(D1):D862–D868PubMedCrossRef
171.
go back to reference Sim NL, Kumar P, Hu J, Henikoff S, Schneider G, Ng PC (2012) SIFT web server: predicting effects of amino acid substitutions on proteins. Nucleic Acids Res 40(Web Server issue):W452–W457PubMedPubMedCentralCrossRef Sim NL, Kumar P, Hu J, Henikoff S, Schneider G, Ng PC (2012) SIFT web server: predicting effects of amino acid substitutions on proteins. Nucleic Acids Res 40(Web Server issue):W452–W457PubMedPubMedCentralCrossRef
172.
go back to reference Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P et al (2010) A method and server for predicting damaging missense mutations. Nat Methods 7(4):248–249PubMedPubMedCentralCrossRef Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P et al (2010) A method and server for predicting damaging missense mutations. Nat Methods 7(4):248–249PubMedPubMedCentralCrossRef
174.
go back to reference Frousios K, Iliopoulos CS, Schlitt T, Simpson MA (2013) Predicting the functional consequences of non-synonymous DNA sequence variants--evaluation of bioinformatics tools and development of a consensus strategy. Genomics 102(4):223–228PubMedCrossRef Frousios K, Iliopoulos CS, Schlitt T, Simpson MA (2013) Predicting the functional consequences of non-synonymous DNA sequence variants--evaluation of bioinformatics tools and development of a consensus strategy. Genomics 102(4):223–228PubMedCrossRef
175.
go back to reference Schwarz JM, Rodelsperger C, Schuelke M, Seelow D (2010) MutationTaster evaluates disease-causing potential of sequence alterations. Nat Methods 7(8):575–576PubMedCrossRef Schwarz JM, Rodelsperger C, Schuelke M, Seelow D (2010) MutationTaster evaluates disease-causing potential of sequence alterations. Nat Methods 7(8):575–576PubMedCrossRef
176.
go back to reference Lee D, Gorkin DU, Baker M, Strober BJ, Asoni AL, McCallion AS et al (2015) A method to predict the impact of regulatory variants from DNA sequence. Nat Genet 47(8):955–961PubMedPubMedCentralCrossRef Lee D, Gorkin DU, Baker M, Strober BJ, Asoni AL, McCallion AS et al (2015) A method to predict the impact of regulatory variants from DNA sequence. Nat Genet 47(8):955–961PubMedPubMedCentralCrossRef
179.
go back to reference Nemeth AH, Kwasniewska AC, Lise S, Parolin Schnekenberg R, Becker EB, Bera KD et al (2013) Next generation sequencing for molecular diagnosis of neurological disorders using ataxias as a model. Brain 136(Pt 10):3106–3118PubMedPubMedCentralCrossRef Nemeth AH, Kwasniewska AC, Lise S, Parolin Schnekenberg R, Becker EB, Bera KD et al (2013) Next generation sequencing for molecular diagnosis of neurological disorders using ataxias as a model. Brain 136(Pt 10):3106–3118PubMedPubMedCentralCrossRef
180.
go back to reference Handel AE, Chintawar S, Lalic T, Whiteley E, Vowles J, Giustacchini A et al (2016) Assessing similarity to primary tissue and cortical layer identity in induced pluripotent stem cell-derived cortical neurons through single-cell transcriptomics. Hum Mol Genet 25(5):989–1000PubMedPubMedCentralCrossRef Handel AE, Chintawar S, Lalic T, Whiteley E, Vowles J, Giustacchini A et al (2016) Assessing similarity to primary tissue and cortical layer identity in induced pluripotent stem cell-derived cortical neurons through single-cell transcriptomics. Hum Mol Genet 25(5):989–1000PubMedPubMedCentralCrossRef
181.
go back to reference Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME et al (2013) Cerebral organoids model human brain development and microcephaly. Nature 501(7467):373–379PubMedCrossRef Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME et al (2013) Cerebral organoids model human brain development and microcephaly. Nature 501(7467):373–379PubMedCrossRef
182.
go back to reference Ipe J, Swart M, Burgess KS, Skaar TC (2017) High-throughput assays to assess the functional impact of genetic variants: a road towards genomic-driven medicine. Clin Transl Sci 10(2):67–77PubMedPubMedCentralCrossRef Ipe J, Swart M, Burgess KS, Skaar TC (2017) High-throughput assays to assess the functional impact of genetic variants: a road towards genomic-driven medicine. Clin Transl Sci 10(2):67–77PubMedPubMedCentralCrossRef
183.
go back to reference Fowler DM, Stephany JJ, Fields S (2014) Measuring the activity of protein variants on a large scale using deep mutational scanning. Nat Protoc 9(9):2267–2284PubMedPubMedCentralCrossRef Fowler DM, Stephany JJ, Fields S (2014) Measuring the activity of protein variants on a large scale using deep mutational scanning. Nat Protoc 9(9):2267–2284PubMedPubMedCentralCrossRef
184.
go back to reference Li T, Lu G, Chiang EY, Chernov-Rogan T, Grogan JL, Chen J (2017) High-throughput electrophysiological assays for voltage gated ion channels using SyncroPatch 768PE. PLoS One 12(7):e0180154PubMedPubMedCentralCrossRef Li T, Lu G, Chiang EY, Chernov-Rogan T, Grogan JL, Chen J (2017) High-throughput electrophysiological assays for voltage gated ion channels using SyncroPatch 768PE. PLoS One 12(7):e0180154PubMedPubMedCentralCrossRef
185.
go back to reference Dale RC, Gardiner A, Branson JA, Houlden H (2014) Benefit of carbamazepine in a patient with hemiplegic migraine associated with PRRT2 mutation. Dev Med Child Neurol 56(9):910PubMedCrossRef Dale RC, Gardiner A, Branson JA, Houlden H (2014) Benefit of carbamazepine in a patient with hemiplegic migraine associated with PRRT2 mutation. Dev Med Child Neurol 56(9):910PubMedCrossRef
186.
go back to reference Maher BH, Griffiths LR (2011) Identification of molecular genetic factors that influence migraine. Mol Gen Genomics 285(6):433–446CrossRef Maher BH, Griffiths LR (2011) Identification of molecular genetic factors that influence migraine. Mol Gen Genomics 285(6):433–446CrossRef
187.
go back to reference Kondratieva N, Azimova J, Skorobogatykh K, Sergeev A, Naumova E, Kokaeva Z et al (2016) Biomarkers of migraine: part 1 - genetic markers. J Neurol Sci 369:63–76PubMedCrossRef Kondratieva N, Azimova J, Skorobogatykh K, Sergeev A, Naumova E, Kokaeva Z et al (2016) Biomarkers of migraine: part 1 - genetic markers. J Neurol Sci 369:63–76PubMedCrossRef
188.
go back to reference Frosst P, Blom HJ, Milos R, Goyette P, Sheppard CA, Matthews RG et al (1995) A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet 10(1):111–113PubMedCrossRef Frosst P, Blom HJ, Milos R, Goyette P, Sheppard CA, Matthews RG et al (1995) A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet 10(1):111–113PubMedCrossRef
189.
go back to reference Rubino E, Ferrero M, Rainero I, Binello E, Vaula G, Pinessi L (2009) Association of the C677T polymorphism in the MTHFR gene with migraine: a meta-analysis. Cephalalgia 29(8):818–825PubMedCrossRef Rubino E, Ferrero M, Rainero I, Binello E, Vaula G, Pinessi L (2009) Association of the C677T polymorphism in the MTHFR gene with migraine: a meta-analysis. Cephalalgia 29(8):818–825PubMedCrossRef
190.
go back to reference Schurks M, Rist PM, Kurth T (2010) MTHFR 677C>T and ACE D/I polymorphisms in migraine: a systematic review and meta-analysis. Headache 50(4):588–599PubMedCrossRef Schurks M, Rist PM, Kurth T (2010) MTHFR 677C>T and ACE D/I polymorphisms in migraine: a systematic review and meta-analysis. Headache 50(4):588–599PubMedCrossRef
191.
go back to reference Samaan Z, Gaysina D, Cohen-Woods S, Craddock N, Jones L, Korszun A et al (2011) Methylenetetrahydrofolate reductase gene variant (MTHFR C677T) and migraine: a case control study and meta-analysis. BMC Neurol 11:66PubMedPubMedCentralCrossRef Samaan Z, Gaysina D, Cohen-Woods S, Craddock N, Jones L, Korszun A et al (2011) Methylenetetrahydrofolate reductase gene variant (MTHFR C677T) and migraine: a case control study and meta-analysis. BMC Neurol 11:66PubMedPubMedCentralCrossRef
192.
go back to reference Liu R, Geng P, Ma M, Yu S, Yang M, He M et al (2014) MTHFR C677T polymorphism and migraine risk: a meta-analysis. J Neurol Sci 336(1–2):68–73PubMedCrossRef Liu R, Geng P, Ma M, Yu S, Yang M, He M et al (2014) MTHFR C677T polymorphism and migraine risk: a meta-analysis. J Neurol Sci 336(1–2):68–73PubMedCrossRef
193.
go back to reference Kaunisto MA, Kallela M, Hamalainen E, Kilpikari R, Havanka H, Harno H et al (2006) Testing of variants of the MTHFR and ESR1 genes in 1798 Finnish individuals fails to confirm the association with migraine with aura. Cephalalgia 26(12):1462–1472PubMedCrossRef Kaunisto MA, Kallela M, Hamalainen E, Kilpikari R, Havanka H, Harno H et al (2006) Testing of variants of the MTHFR and ESR1 genes in 1798 Finnish individuals fails to confirm the association with migraine with aura. Cephalalgia 26(12):1462–1472PubMedCrossRef
194.
go back to reference Todt U, Freudenberg J, Goebel I, Netzer C, Heinze A, Heinze-Kuhn K et al (2006) MTHFR C677T polymorphism and migraine with aura. Ann Neurol 60(5):621–622 author reply 2-3PubMedCrossRef Todt U, Freudenberg J, Goebel I, Netzer C, Heinze A, Heinze-Kuhn K et al (2006) MTHFR C677T polymorphism and migraine with aura. Ann Neurol 60(5):621–622 author reply 2-3PubMedCrossRef
195.
go back to reference de Vries B, Anttila V, Freilinger T, Wessman M, Kaunisto MA, Kallela M et al (2016) Systematic re-evaluation of genes from candidate gene association studies in migraine using a large genome-wide association data set. Cephalalgia 36(7):604–614PubMedCrossRef de Vries B, Anttila V, Freilinger T, Wessman M, Kaunisto MA, Kallela M et al (2016) Systematic re-evaluation of genes from candidate gene association studies in migraine using a large genome-wide association data set. Cephalalgia 36(7):604–614PubMedCrossRef
196.
go back to reference Cardon LR, Palmer LJ (2003) Population stratification and spurious allelic association. Lancet 361(9357):598–604PubMedCrossRef Cardon LR, Palmer LJ (2003) Population stratification and spurious allelic association. Lancet 361(9357):598–604PubMedCrossRef
197.
go back to reference Anttila V, Stefansson H, Kallela M, Todt U, Terwindt GM, Calafato MS et al (2010) Genome-wide association study of migraine implicates a common susceptibility variant on 8q22. Nat Genet 42(10):869–873PubMedPubMedCentralCrossRef Anttila V, Stefansson H, Kallela M, Todt U, Terwindt GM, Calafato MS et al (2010) Genome-wide association study of migraine implicates a common susceptibility variant on 8q22. Nat Genet 42(10):869–873PubMedPubMedCentralCrossRef
198.
go back to reference Chasman DI, Schurks M, Anttila V, de Vries B, Schminke U, Launer LJ et al (2011) Genome-wide association study reveals three susceptibility loci for common migraine in the general population. Nat Genet 43(7):695–698PubMedPubMedCentralCrossRef Chasman DI, Schurks M, Anttila V, de Vries B, Schminke U, Launer LJ et al (2011) Genome-wide association study reveals three susceptibility loci for common migraine in the general population. Nat Genet 43(7):695–698PubMedPubMedCentralCrossRef
199.
go back to reference Freilinger T, Anttila V, de Vries B, Malik R, Kallela M, Terwindt GM et al (2012) Genome-wide association analysis identifies susceptibility loci for migraine without aura. Nat Genet 44(7):777–782PubMedPubMedCentralCrossRef Freilinger T, Anttila V, de Vries B, Malik R, Kallela M, Terwindt GM et al (2012) Genome-wide association analysis identifies susceptibility loci for migraine without aura. Nat Genet 44(7):777–782PubMedPubMedCentralCrossRef
200.
go back to reference Anttila V, Winsvold BS, Gormley P, Kurth T, Bettella F, McMahon G et al (2013) Genome-wide meta-analysis identifies new susceptibility loci for migraine. Nat Genet 45(8):912–917PubMedPubMedCentralCrossRef Anttila V, Winsvold BS, Gormley P, Kurth T, Bettella F, McMahon G et al (2013) Genome-wide meta-analysis identifies new susceptibility loci for migraine. Nat Genet 45(8):912–917PubMedPubMedCentralCrossRef
201.
go back to reference Gan M, Jiang P, McLean P, Kanekiyo T, Bu G (2014) Low-density lipoprotein receptor-related protein 1 (LRP1) regulates the stability and function of GluA1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor in neurons. PLoS One 9(12):e113237PubMedPubMedCentralCrossRef Gan M, Jiang P, McLean P, Kanekiyo T, Bu G (2014) Low-density lipoprotein receptor-related protein 1 (LRP1) regulates the stability and function of GluA1 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor in neurons. PLoS One 9(12):e113237PubMedPubMedCentralCrossRef
202.
go back to reference Spuch C, Ortolano S, Navarro C (2012) LRP-1 and LRP-2 receptors function in the membrane neuron. Trafficking mechanisms and proteolytic processing in Alzheimer's disease. Front Physiol 3:269PubMedPubMedCentralCrossRef Spuch C, Ortolano S, Navarro C (2012) LRP-1 and LRP-2 receptors function in the membrane neuron. Trafficking mechanisms and proteolytic processing in Alzheimer's disease. Front Physiol 3:269PubMedPubMedCentralCrossRef
203.
go back to reference Nakajima C, Kulik A, Frotscher M, Herz J, Schafer M, Bock HH et al (2013) Low density lipoprotein receptor-related protein 1 (LRP1) modulates N-methyl-D-aspartate (NMDA) receptor-dependent intracellular signaling and NMDA-induced regulation of postsynaptic protein complexes. J Biol Chem 288(30):21909–21923PubMedPubMedCentralCrossRef Nakajima C, Kulik A, Frotscher M, Herz J, Schafer M, Bock HH et al (2013) Low density lipoprotein receptor-related protein 1 (LRP1) modulates N-methyl-D-aspartate (NMDA) receptor-dependent intracellular signaling and NMDA-induced regulation of postsynaptic protein complexes. J Biol Chem 288(30):21909–21923PubMedPubMedCentralCrossRef
205.
go back to reference Chi J, Cohen P (2016) The multifaceted roles of PRDM16: adipose biology and beyond. Trends Endocrinol Metab 27(1):11–23PubMedCrossRef Chi J, Cohen P (2016) The multifaceted roles of PRDM16: adipose biology and beyond. Trends Endocrinol Metab 27(1):11–23PubMedCrossRef
206.
go back to reference Chuikov S, Levi BP, Smith ML, Morrison SJ (2010) Prdm16 promotes stem cell maintenance in multiple tissues, partly by regulating oxidative stress. Nat Cell Biol 12(10):999–1006PubMedPubMedCentralCrossRef Chuikov S, Levi BP, Smith ML, Morrison SJ (2010) Prdm16 promotes stem cell maintenance in multiple tissues, partly by regulating oxidative stress. Nat Cell Biol 12(10):999–1006PubMedPubMedCentralCrossRef
207.
go back to reference Shimada IS, Acar M, Burgess RJ, Zhao Z, Morrison SJ (2017) Prdm16 is required for the maintenance of neural stem cells in the postnatal forebrain and their differentiation into ependymal cells. Genes Dev 31(11):1134–1146PubMedPubMedCentralCrossRef Shimada IS, Acar M, Burgess RJ, Zhao Z, Morrison SJ (2017) Prdm16 is required for the maintenance of neural stem cells in the postnatal forebrain and their differentiation into ependymal cells. Genes Dev 31(11):1134–1146PubMedPubMedCentralCrossRef
208.
go back to reference Nguyen HL, Lee YJ, Shin J, Lee E, Park SO, McCarty JH et al (2011) TGF-beta signaling in endothelial cells, but not neuroepithelial cells, is essential for cerebral vascular development. Lab Investig 91(11):1554–1563PubMedCrossRef Nguyen HL, Lee YJ, Shin J, Lee E, Park SO, McCarty JH et al (2011) TGF-beta signaling in endothelial cells, but not neuroepithelial cells, is essential for cerebral vascular development. Lab Investig 91(11):1554–1563PubMedCrossRef
209.
go back to reference Luo SX, Timbang L, Kim JI, Shang Y, Sandoval K, Tang AA et al (2016) TGF-beta signaling in dopaminergic neurons regulates dendritic growth, excitatory-inhibitory synaptic balance, and reversal learning. Cell Rep 17(12):3233–3245PubMedPubMedCentralCrossRef Luo SX, Timbang L, Kim JI, Shang Y, Sandoval K, Tang AA et al (2016) TGF-beta signaling in dopaminergic neurons regulates dendritic growth, excitatory-inhibitory synaptic balance, and reversal learning. Cell Rep 17(12):3233–3245PubMedPubMedCentralCrossRef
211.
go back to reference McAllister BB, Dyck RH (2017) Zinc transporter 3 (ZnT3) and vesicular zinc in central nervous system function. Neurosci Biobehav Rev 80:329–350PubMedCrossRef McAllister BB, Dyck RH (2017) Zinc transporter 3 (ZnT3) and vesicular zinc in central nervous system function. Neurosci Biobehav Rev 80:329–350PubMedCrossRef
212.
go back to reference Telley L, Cadilhac C, Cioni JM, Saywell V, Jahannault-Talignani C, Huettl RE et al (2016) Dual function of NRP1 in axon guidance and subcellular target recognition in cerebellum. Neuron 91(6):1276–1291PubMedCrossRef Telley L, Cadilhac C, Cioni JM, Saywell V, Jahannault-Talignani C, Huettl RE et al (2016) Dual function of NRP1 in axon guidance and subcellular target recognition in cerebellum. Neuron 91(6):1276–1291PubMedCrossRef
213.
go back to reference Aghajanian H, Cho YK, Manderfield LJ, Herling MR, Gupta M, Ho VC et al (2016) Coronary vasculature patterning requires a novel endothelial ErbB2 holoreceptor. Nat Commun 7:12038PubMedPubMedCentralCrossRef Aghajanian H, Cho YK, Manderfield LJ, Herling MR, Gupta M, Ho VC et al (2016) Coronary vasculature patterning requires a novel endothelial ErbB2 holoreceptor. Nat Commun 7:12038PubMedPubMedCentralCrossRef
215.
go back to reference Wellcome Trust Case Control C, Maller JB, McVean G, Byrnes J, Vukcevic D, Palin K et al (2012) Bayesian refinement of association signals for 14 loci in 3 common diseases. Nat Genet 44(12):1294–1301CrossRef Wellcome Trust Case Control C, Maller JB, McVean G, Byrnes J, Vukcevic D, Palin K et al (2012) Bayesian refinement of association signals for 14 loci in 3 common diseases. Nat Genet 44(12):1294–1301CrossRef
216.
go back to reference Gupta RM, Hadaya J, Trehan A, Zekavat SM, Roselli C, Klarin D et al (2017) A genetic variant associated with five vascular diseases is a distal regulator of Endothelin-1 gene expression. Cell 170(3):522–33 e15PubMedPubMedCentralCrossRef Gupta RM, Hadaya J, Trehan A, Zekavat SM, Roselli C, Klarin D et al (2017) A genetic variant associated with five vascular diseases is a distal regulator of Endothelin-1 gene expression. Cell 170(3):522–33 e15PubMedPubMedCentralCrossRef
217.
go back to reference Hannon E, Weedon M, Bray N, O'Donovan M, Mill J (2017) Pleiotropic effects of trait-associated genetic variation on DNA methylation: utility for refining GWAS loci. Am J Hum Genet 100(6):954–959PubMedPubMedCentralCrossRef Hannon E, Weedon M, Bray N, O'Donovan M, Mill J (2017) Pleiotropic effects of trait-associated genetic variation on DNA methylation: utility for refining GWAS loci. Am J Hum Genet 100(6):954–959PubMedPubMedCentralCrossRef
218.
go back to reference Eising E, Huisman SM, Mahfouz A, Vijfhuizen LS, Anttila V, Winsvold BS et al (2016) Gene co-expression analysis identifies brain regions and cell types involved in migraine pathophysiology: a GWAS-based study using the Allen human brain atlas. Hum Genet 135(4):425–439PubMedPubMedCentralCrossRef Eising E, Huisman SM, Mahfouz A, Vijfhuizen LS, Anttila V, Winsvold BS et al (2016) Gene co-expression analysis identifies brain regions and cell types involved in migraine pathophysiology: a GWAS-based study using the Allen human brain atlas. Hum Genet 135(4):425–439PubMedPubMedCentralCrossRef
219.
go back to reference Renthal W (2018) Localization of migraine susceptibility genes in human brain by single-cell RNA sequencing. Cephalalgia 38(13):1976–1983PubMedCrossRef Renthal W (2018) Localization of migraine susceptibility genes in human brain by single-cell RNA sequencing. Cephalalgia 38(13):1976–1983PubMedCrossRef
220.
go back to reference LaPaglia DM, Sapio MR, Burbelo PD, Thierry-Mieg J, Thierry-Mieg D, Raithel SJ et al (2018) RNA-Seq investigations of human post-mortem trigeminal ganglia. Cephalalgia 38(5):912–932PubMedCrossRef LaPaglia DM, Sapio MR, Burbelo PD, Thierry-Mieg J, Thierry-Mieg D, Raithel SJ et al (2018) RNA-Seq investigations of human post-mortem trigeminal ganglia. Cephalalgia 38(5):912–932PubMedCrossRef
221.
go back to reference Russell MB, Ulrich V, Gervil M, Olesen J (2002) Migraine without aura and migraine with aura are distinct disorders. A population-based twin survey. Headache 42(5):332–336PubMedCrossRef Russell MB, Ulrich V, Gervil M, Olesen J (2002) Migraine without aura and migraine with aura are distinct disorders. A population-based twin survey. Headache 42(5):332–336PubMedCrossRef
222.
go back to reference Nyholt DR, Gillespie NG, Heath AC, Merikangas KR, Duffy DL, Martin NG (2004) Latent class and genetic analysis does not support migraine with aura and migraine without aura as separate entities. Genet Epidemiol 26(3):231–244PubMedCrossRef Nyholt DR, Gillespie NG, Heath AC, Merikangas KR, Duffy DL, Martin NG (2004) Latent class and genetic analysis does not support migraine with aura and migraine without aura as separate entities. Genet Epidemiol 26(3):231–244PubMedCrossRef
223.
go back to reference Zhao H, Eising E, de Vries B, Vijfhuizen LS (2016) International headache genetics C, Anttila V, et al. gene-based pleiotropy across migraine with aura and migraine without aura patient groups. Cephalalgia 36(7):648–657PubMedCrossRef Zhao H, Eising E, de Vries B, Vijfhuizen LS (2016) International headache genetics C, Anttila V, et al. gene-based pleiotropy across migraine with aura and migraine without aura patient groups. Cephalalgia 36(7):648–657PubMedCrossRef
224.
go back to reference Chasman DI, Anttila V, Buring JE, Ridker PM, Schurks M, Kurth T et al (2014) Selectivity in genetic association with sub-classified migraine in women. PLoS Genet 10(5):e1004366PubMedPubMedCentralCrossRef Chasman DI, Anttila V, Buring JE, Ridker PM, Schurks M, Kurth T et al (2014) Selectivity in genetic association with sub-classified migraine in women. PLoS Genet 10(5):e1004366PubMedPubMedCentralCrossRef
225.
go back to reference Esserlind AL, Christensen AF, Steinberg S, Grarup N, Pedersen O, Hansen T et al (2016) The association between candidate migraine susceptibility loci and severe migraine phenotype in a clinical sample. Cephalalgia 36(7):615–623PubMedCrossRef Esserlind AL, Christensen AF, Steinberg S, Grarup N, Pedersen O, Hansen T et al (2016) The association between candidate migraine susceptibility loci and severe migraine phenotype in a clinical sample. Cephalalgia 36(7):615–623PubMedCrossRef
226.
go back to reference Pollock CE, Sutherland HG, Maher BH, Lea RA, Haupt LM, Frith A et al (2018) The NRP1 migraine risk variant shows evidence of association with menstrual migraine. J Headache Pain 19(1):31PubMedPubMedCentralCrossRef Pollock CE, Sutherland HG, Maher BH, Lea RA, Haupt LM, Frith A et al (2018) The NRP1 migraine risk variant shows evidence of association with menstrual migraine. J Headache Pain 19(1):31PubMedPubMedCentralCrossRef
227.
go back to reference Meng W, Adams MJ, Hebert HL, Deary IJ, McIntosh AM, Smith BH (2018) A genome-wide association study finds genetic associations with broadly-defined headache in UK biobank (N=223,773). EBioMedicine 28:180–186PubMedPubMedCentralCrossRef Meng W, Adams MJ, Hebert HL, Deary IJ, McIntosh AM, Smith BH (2018) A genome-wide association study finds genetic associations with broadly-defined headache in UK biobank (N=223,773). EBioMedicine 28:180–186PubMedPubMedCentralCrossRef
228.
go back to reference Brainstorm Consortium, Anttila V, Bulik-Sullivan B, Finucane HK, Walters RK, Bras J et al (2018) Analysis of shared heritability in common disorders of the brain. Science 360(6395):eaap8757 Brainstorm Consortium, Anttila V, Bulik-Sullivan B, Finucane HK, Walters RK, Bras J et al (2018) Analysis of shared heritability in common disorders of the brain. Science 360(6395):eaap8757
229.
go back to reference Yang Y, Zhao H, Boomsma DI, Ligthart L, Belin AC, Smith GD et al (2018) Molecular genetic overlap between migraine and major depressive disorder. Eur J Hum Genet 26(8):1202–1216PubMedCrossRefPubMedCentral Yang Y, Zhao H, Boomsma DI, Ligthart L, Belin AC, Smith GD et al (2018) Molecular genetic overlap between migraine and major depressive disorder. Eur J Hum Genet 26(8):1202–1216PubMedCrossRefPubMedCentral
230.
go back to reference Keezer MR, Bauer PR, Ferrari MD, Sander JW (2015) The comorbid relationship between migraine and epilepsy: a systematic review and meta-analysis. Eur J Neurol 22(7):1038–1047PubMedCrossRef Keezer MR, Bauer PR, Ferrari MD, Sander JW (2015) The comorbid relationship between migraine and epilepsy: a systematic review and meta-analysis. Eur J Neurol 22(7):1038–1047PubMedCrossRef
231.
go back to reference Malik R, Freilinger T, Winsvold BS, Anttila V, Vander Heiden J, Traylor M et al (2015) Shared genetic basis for migraine and ischemic stroke: a genome-wide analysis of common variants. Neurology 84(21):2132–2145PubMedPubMedCentralCrossRef Malik R, Freilinger T, Winsvold BS, Anttila V, Vander Heiden J, Traylor M et al (2015) Shared genetic basis for migraine and ischemic stroke: a genome-wide analysis of common variants. Neurology 84(21):2132–2145PubMedPubMedCentralCrossRef
232.
go back to reference Winsvold BS, Nelson CP, Malik R, Gormley P, Anttila V, Vander Heiden J et al (2015) Genetic analysis for a shared biological basis between migraine and coronary artery disease. Neurol Genet 1(1):e10PubMedPubMedCentralCrossRef Winsvold BS, Nelson CP, Malik R, Gormley P, Anttila V, Vander Heiden J et al (2015) Genetic analysis for a shared biological basis between migraine and coronary artery disease. Neurol Genet 1(1):e10PubMedPubMedCentralCrossRef
233.
go back to reference Kutuk MO, Tufan AE, Guler G, Yalin OO, Altintas E, Bag HG et al (2018) Migraine and associated comorbidities are three times more frequent in children with ADHD and their mothers. Brain and Development 40(10):857–864PubMedCrossRef Kutuk MO, Tufan AE, Guler G, Yalin OO, Altintas E, Bag HG et al (2018) Migraine and associated comorbidities are three times more frequent in children with ADHD and their mothers. Brain and Development 40(10):857–864PubMedCrossRef
234.
go back to reference Prontera P, Sarchielli P, Caproni S, Bedetti C, Cupini LM, Calabresi P et al (2018) Epilepsy in hemiplegic migraine: genetic mutations and clinical implications. Cephalalgia 38(2):361–373PubMedCrossRef Prontera P, Sarchielli P, Caproni S, Bedetti C, Cupini LM, Calabresi P et al (2018) Epilepsy in hemiplegic migraine: genetic mutations and clinical implications. Cephalalgia 38(2):361–373PubMedCrossRef
235.
go back to reference Louter MA, Pelzer N, de Boer I, Kuijvenhoven BE, van Oosterhout WP, van Zwet EW et al (2016) Prevalence of lifetime depression in a large hemiplegic migraine cohort. Neurology 87(22):2370–2374PubMedCrossRef Louter MA, Pelzer N, de Boer I, Kuijvenhoven BE, van Oosterhout WP, van Zwet EW et al (2016) Prevalence of lifetime depression in a large hemiplegic migraine cohort. Neurology 87(22):2370–2374PubMedCrossRef
236.
go back to reference Linde M, Mulleners WM, Chronicle EP, McCrory DC (2013) Valproate (valproic acid or sodium valproate or a combination of the two) for the prophylaxis of episodic migraine in adults. Cochrane Database Syst Rev 24(6):CD010611 Linde M, Mulleners WM, Chronicle EP, McCrory DC (2013) Valproate (valproic acid or sodium valproate or a combination of the two) for the prophylaxis of episodic migraine in adults. Cochrane Database Syst Rev 24(6):CD010611
237.
go back to reference Ran C, Graae L, Magnusson PK, Pedersen NL, Olson L, Belin AC (2014) A replication study of GWAS findings in migraine identifies association in a Swedish case-control sample. BMC Med Genet 15:38PubMedPubMedCentralCrossRef Ran C, Graae L, Magnusson PK, Pedersen NL, Olson L, Belin AC (2014) A replication study of GWAS findings in migraine identifies association in a Swedish case-control sample. BMC Med Genet 15:38PubMedPubMedCentralCrossRef
238.
go back to reference Sintas C, Fernandez-Morales J, Vila-Pueyo M, Narberhaus B, Arenas C, Pozo-Rosich P et al (2015) Replication study of previous migraine genome-wide association study findings in a Spanish sample of migraine with aura. Cephalalgia 35(9):776–782PubMedCrossRef Sintas C, Fernandez-Morales J, Vila-Pueyo M, Narberhaus B, Arenas C, Pozo-Rosich P et al (2015) Replication study of previous migraine genome-wide association study findings in a Spanish sample of migraine with aura. Cephalalgia 35(9):776–782PubMedCrossRef
239.
go back to reference An XK, Ma QL, Lin Q, Zhang XR, Lu CX, Qu HL (2013) PRDM16 rs2651899 variant is a risk factor for Chinese common migraine patients. Headache 53(10):1595–1601PubMedCrossRef An XK, Ma QL, Lin Q, Zhang XR, Lu CX, Qu HL (2013) PRDM16 rs2651899 variant is a risk factor for Chinese common migraine patients. Headache 53(10):1595–1601PubMedCrossRef
240.
go back to reference Fan X, Wang J, Fan W, Chen L, Gui B, Tan G et al (2014) Replication of migraine GWAS susceptibility loci in Chinese Han population. Headache 54(4):709–715PubMedCrossRef Fan X, Wang J, Fan W, Chen L, Gui B, Tan G et al (2014) Replication of migraine GWAS susceptibility loci in Chinese Han population. Headache 54(4):709–715PubMedCrossRef
241.
go back to reference Ghosh J, Pradhan S, Mittal B (2013) Genome-wide-associated variants in migraine susceptibility: a replication study from North India. Headache 53(10):1583–1594PubMedCrossRef Ghosh J, Pradhan S, Mittal B (2013) Genome-wide-associated variants in migraine susceptibility: a replication study from North India. Headache 53(10):1583–1594PubMedCrossRef
242.
go back to reference Lin QF, Fu XG, Yao LT, Yang J, Cao LY, Xin YT et al (2015) Association of genetic loci for migraine susceptibility in the she people of China. J Headache Pain 16:553PubMedCrossRef Lin QF, Fu XG, Yao LT, Yang J, Cao LY, Xin YT et al (2015) Association of genetic loci for migraine susceptibility in the she people of China. J Headache Pain 16:553PubMedCrossRef
243.
go back to reference Chen SP, Fuh JL, Chung MY, Lin YC, Liao YC, Wang YF et al (2018) Genome-wide association study identifies novel susceptibility loci for migraine in Han Chinese resided in Taiwan. Cephalalgia 38(3):466–475PubMedCrossRef Chen SP, Fuh JL, Chung MY, Lin YC, Liao YC, Wang YF et al (2018) Genome-wide association study identifies novel susceptibility loci for migraine in Han Chinese resided in Taiwan. Cephalalgia 38(3):466–475PubMedCrossRef
244.
go back to reference Chang X, Pellegrino R, Garifallou J, March M, Snyder J, Mentch F et al (2018) Common variants at 5q33.1 predispose to migraine in African-American children. J Med Genet 55(12):831–836PubMedCrossRef Chang X, Pellegrino R, Garifallou J, March M, Snyder J, Mentch F et al (2018) Common variants at 5q33.1 predispose to migraine in African-American children. J Med Genet 55(12):831–836PubMedCrossRef
245.
go back to reference Cox HC, Lea RA, Bellis C, Carless M, Dyer TD, Curran J et al (2012) A genome-wide analysis of 'Bounty' descendants implicates several novel variants in migraine susceptibility. Neurogenetics 13(3):261–266PubMedPubMedCentralCrossRef Cox HC, Lea RA, Bellis C, Carless M, Dyer TD, Curran J et al (2012) A genome-wide analysis of 'Bounty' descendants implicates several novel variants in migraine susceptibility. Neurogenetics 13(3):261–266PubMedPubMedCentralCrossRef
248.
go back to reference Rodriguez-Acevedo AJ, Ferreira MA, Benton MC, Carless MA, Goring HH, Curran JE et al (2015) Common polygenic variation contributes to risk of migraine in the Norfolk Island population. Hum Genet 134(10):1079–1087PubMedCrossRef Rodriguez-Acevedo AJ, Ferreira MA, Benton MC, Carless MA, Goring HH, Curran JE et al (2015) Common polygenic variation contributes to risk of migraine in the Norfolk Island population. Hum Genet 134(10):1079–1087PubMedCrossRef
249.
go back to reference Gormley P, Kurki MI, Hiekkala ME, Veerapen K, Happola P, Mitchell AA et al (2018) Common variant burden contributes to the familial aggregation of migraine in 1,589 families. Neuron 98(4):743–53 e4PubMedPubMedCentralCrossRef Gormley P, Kurki MI, Hiekkala ME, Veerapen K, Happola P, Mitchell AA et al (2018) Common variant burden contributes to the familial aggregation of migraine in 1,589 families. Neuron 98(4):743–53 e4PubMedPubMedCentralCrossRef
250.
go back to reference Christensen AF, Esserlind AL, Werge T, Stefansson H, Stefansson K, Olesen J (2016) The influence of genetic constitution on migraine drug responses. Cephalalgia 36(7):624–639PubMedCrossRef Christensen AF, Esserlind AL, Werge T, Stefansson H, Stefansson K, Olesen J (2016) The influence of genetic constitution on migraine drug responses. Cephalalgia 36(7):624–639PubMedCrossRef
251.
go back to reference Cargnin S, Viana M, Sances G, Cantello R, Tassorelli C, Terrazzino S (2019) Using a genetic risk score approach to predict headache response to Triptans in migraine without Aura. J Clin Pharmacol 59(2):288–294PubMedCrossRef Cargnin S, Viana M, Sances G, Cantello R, Tassorelli C, Terrazzino S (2019) Using a genetic risk score approach to predict headache response to Triptans in migraine without Aura. J Clin Pharmacol 59(2):288–294PubMedCrossRef
252.
go back to reference Schurks M, Kurth T, Stude P, Rimmbach C, de Jesus J, Jonjic M et al (2007) G protein beta3 polymorphism and triptan response in cluster headache. Clin Pharmacol Ther 82(4):396–401PubMedCrossRef Schurks M, Kurth T, Stude P, Rimmbach C, de Jesus J, Jonjic M et al (2007) G protein beta3 polymorphism and triptan response in cluster headache. Clin Pharmacol Ther 82(4):396–401PubMedCrossRef
253.
go back to reference Gentile G, Missori S, Borro M, Sebastianelli A, Simmaco M, Martelletti P (2010) Frequencies of genetic polymorphisms related to triptans metabolism in chronic migraine. J Headache Pain 11(2):151–156PubMedPubMedCentralCrossRef Gentile G, Missori S, Borro M, Sebastianelli A, Simmaco M, Martelletti P (2010) Frequencies of genetic polymorphisms related to triptans metabolism in chronic migraine. J Headache Pain 11(2):151–156PubMedPubMedCentralCrossRef
254.
go back to reference Wood AR, Esko T, Yang J, Vedantam S, Pers TH, Gustafsson S et al (2014) Defining the role of common variation in the genomic and biological architecture of adult human height. Nat Genet 46(11):1173–1186PubMedPubMedCentralCrossRef Wood AR, Esko T, Yang J, Vedantam S, Pers TH, Gustafsson S et al (2014) Defining the role of common variation in the genomic and biological architecture of adult human height. Nat Genet 46(11):1173–1186PubMedPubMedCentralCrossRef
255.
go back to reference Zhong K, Zhu G, Jing X, Hendriks AEJ, Drop SLS, Ikram MA et al (2017) Genome-wide compound heterozygote analysis highlights alleles associated with adult height in Europeans. Hum Genet 136(11–12):1407–1417PubMedPubMedCentralCrossRef Zhong K, Zhu G, Jing X, Hendriks AEJ, Drop SLS, Ikram MA et al (2017) Genome-wide compound heterozygote analysis highlights alleles associated with adult height in Europeans. Hum Genet 136(11–12):1407–1417PubMedPubMedCentralCrossRef
256.
go back to reference Marouli E, Graff M, Medina-Gomez C, Lo KS, Wood AR, Kjaer TR et al (2017) Rare and low-frequency coding variants alter human adult height. Nature 542(7640):186–190PubMedPubMedCentralCrossRef Marouli E, Graff M, Medina-Gomez C, Lo KS, Wood AR, Kjaer TR et al (2017) Rare and low-frequency coding variants alter human adult height. Nature 542(7640):186–190PubMedPubMedCentralCrossRef
257.
go back to reference Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR et al (2015) Genetic studies of body mass index yield new insights for obesity biology. Nature 518(7538):197–206PubMedPubMedCentralCrossRef Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR et al (2015) Genetic studies of body mass index yield new insights for obesity biology. Nature 518(7538):197–206PubMedPubMedCentralCrossRef
258.
go back to reference Akiyama M, Okada Y, Kanai M, Takahashi A, Momozawa Y, Ikeda M et al (2017) Genome-wide association study identifies 112 new loci for body mass index in the Japanese population. Nat Genet 49(10):1458–1467PubMedCrossRef Akiyama M, Okada Y, Kanai M, Takahashi A, Momozawa Y, Ikeda M et al (2017) Genome-wide association study identifies 112 new loci for body mass index in the Japanese population. Nat Genet 49(10):1458–1467PubMedCrossRef
259.
go back to reference Gerring ZF, McRae AF, Montgomery GW, Nyholt DR (2018) Genome-wide DNA methylation profiling in whole blood reveals epigenetic signatures associated with migraine. BMC Genomics 19(1):69 Gerring ZF, McRae AF, Montgomery GW, Nyholt DR (2018) Genome-wide DNA methylation profiling in whole blood reveals epigenetic signatures associated with migraine. BMC Genomics 19(1):69
Metadata
Title
Advances in genetics of migraine
Authors
Heidi G. Sutherland
Cassie L. Albury
Lyn R. Griffiths
Publication date
01-12-2019
Publisher
Springer Milan
Keywords
Migraine
Aura
Published in
The Journal of Headache and Pain / Issue 1/2019
Print ISSN: 1129-2369
Electronic ISSN: 1129-2377
DOI
https://doi.org/10.1186/s10194-019-1017-9

Other articles of this Issue 1/2019

The Journal of Headache and Pain 1/2019 Go to the issue