Skip to main content

Advertisement

Log in

Migraine genetics: current findings and future lines of research

  • Review article
  • Published:
neurogenetics Aims and scope Submit manuscript

Abstract

In the last two decades, migraine research has greatly advanced our current knowledge of the genetic contributions and the pathophysiology of this common and debilitating disorder. Nonetheless, this knowledge still needs to grow further and to translate into more effective treatments. To date, several genes involved in syndromic and monogenic forms of migraine have been identified, allowing the generation of animal models which have significantly contributed to current knowledge of the mechanisms underlying these rare forms of migraine. Common forms of migraine are instead posing a greater challenge, as they may most often stem from complex interactions between multiple common genetic variants, with environmental triggers. This paper reviews our current understanding of migraine genetics, moving from syndromic and monogenic forms to oligogenic/polygenic migraines most recently addressed with some success through genome-wide association studies. Methodological issues in study design and future perspectives opened by biomarker research will also be briefly addressed.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Headache Classification Committee of the International Headache Society (IHS) (2013) The international classification of headache disorders, 3rd edition (beta version). Cephalalgia 33:629–808

    Google Scholar 

  2. Bigal ME, Lipton RB (2009) The epidemiology, burden, and comorbidities of migraine. Neurol Clin 27:321–334

    PubMed  Google Scholar 

  3. Robbins MS, Lipton RB (2010) The epidemiology of primary headache disorders. Semin Neurol 30:107–109

    PubMed  Google Scholar 

  4. Noseda R, Burstein R (2013) Migraine pathophysiology: anatomy of the trigeminovascular pathway and associated neurological symptoms, CSD, sensitization and modulation of pain. Pain 154(Suppl 1)

  5. Eikermann-Haerter K, Moskowitz MA (2008) Animal models of migraine headache and aura. Curr Opin Neurol 21:294–300

    PubMed  Google Scholar 

  6. Mulder EJ, van Baal C, Gaist D et al (2003) Genetic and environmental influences on migraine: a twin study across six countries. Twin Res 6:422–431

    PubMed  Google Scholar 

  7. Svensson DA, Larsson B, Waldenlind E, Pedersen NL (2003) Shared rearing environment in migraine: results from twin reared apart and twin reared together. Headache 43:235–244

    PubMed  Google Scholar 

  8. Cologno D, Pascale AD, Manzoni GC (2003) Familial occurrence of migraine with aura in a population-based study. Headache 43:231–234

    CAS  PubMed  Google Scholar 

  9. Schürks M, Rist PM, Kurth T (2010) Sex hormone receptor gene polymorphisms and migraine: a systematic review and meta-analysis. Cephalalgia 30:1306–1328

    PubMed Central  PubMed  Google Scholar 

  10. Russel MB, Iselius L, Olesen J (1996) Migraine without aura and migraine with aura are inherited disorders. Cephalalgia 16:305–309

    Google Scholar 

  11. Lemos C, Alonso I, Barros J et al (2012) Assessing risk factors for migraine: differences in gender transmission. PLoS One 7:e50626

    PubMed Central  CAS  PubMed  Google Scholar 

  12. Peltonen L, GenomEUtwin (2003) GenomEUtwin: a strategy to identify genetic influences on health and disease. Twin Res 6:354–360

    PubMed  Google Scholar 

  13. Friedman DI, De ver Dye T (2009) Migraine and the environment. Headache 49:941–952

    PubMed  Google Scholar 

  14. Eising E, Datson NA, van den Maagdenberg AMJM, Ferrari MD (2013) Epigenetic mechanisms in migraine: a promising avenue? BMC Med 11:26–32

    PubMed Central  PubMed  Google Scholar 

  15. Diener HC, Kuper M, Kurth T (2008) Migraine-associated risks and comorbidity. J Neurol 255:1290–1301

    CAS  PubMed  Google Scholar 

  16. Bellini B, Arruda M, Cescut A et al (2013) Headache and comorbidity in children and adolescents. J Headache Pain 14:79–86

    PubMed Central  PubMed  Google Scholar 

  17. Hung CI, Liu CY, Juang YY, Wang SJ (2006) The impact of migraine on patients with major depressive disorder. Headache 46:469–477

    PubMed  Google Scholar 

  18. Tietjen GE, Brandes JL, Digre KB et al (2007) High prevalence of somatic symptoms and depression in women with disabling chronic headache. Neurology 68:134–140

    CAS  PubMed  Google Scholar 

  19. Joutel A, Corpechot C, Ducros A et al (1996) Notch-3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature 383:707–710

    CAS  PubMed  Google Scholar 

  20. Federico A, Bianchi S, Dotti MT (2005) The spectrum of mutations for CADASIL diagnosis. Neurol Sci 26:117–124

    CAS  PubMed  Google Scholar 

  21. Ungaro C, Mazzei R, Conforti FL et al (2009) CADASIL: extended polymorphisms and mutational analysis of the NOTCH3 gene. J Neurosci Res 87:1162–1167

    CAS  PubMed  Google Scholar 

  22. Mosca L, Marazzi R, Ciccone A et al (2011) NOTCH3 gene mutations in subjects clinically suspected of CADASIL. J Neurol Sci 307:144–148

    CAS  PubMed  Google Scholar 

  23. Ragno M, Fabrizi GM, Cacchiò G et al (2006) Two novel Italian CADASIL families from Central Italy with mutation CGC-TGC at codon 1006 in the exon 19 Notch3 gene. Neurol Sci 27:252–256

    CAS  PubMed  Google Scholar 

  24. Valenti R, Bianchi S, Pescini F et al (2011) First report of a pathogenic mutation on exon 24 of the NOTCH3 gene in a CADASIL family. J Neurol 258:1632–1636

    CAS  PubMed  Google Scholar 

  25. Razvi SS, Davidson R, Bone I, Muir KW (2005) The prevalence of cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy (CADASIL) in the west of Scotland. J Neurol Neurosurg Psychiatry 76:739–741

    PubMed Central  CAS  PubMed  Google Scholar 

  26. Narayan SK, Gorman G, Kalaria RN et al (2012) The minimum prevalence of CADASIL in Northeast England. Neurology 78:1025–1027

    PubMed Central  CAS  PubMed  Google Scholar 

  27. Iso T, Hamamori Y, Kedes L (2003) Notch signaling in vascular development. Arterioscler Thromb Vasc Biol 23:543–553

    CAS  PubMed  Google Scholar 

  28. Alva JA, Iruela-Arispe ML (2004) Notch signaling in vascular morphogenesis. Curr Opin Hematol 11:278–283

    CAS  PubMed  Google Scholar 

  29. Ishiko A, Shimizu A, Nagata E et al (2006) Notch3 ectodomain is a major component of granular osmiophilic material (GOM) in CADASIL. Acta Neuropathol 112:333–339

    CAS  PubMed  Google Scholar 

  30. Liem MK, Oberstein SA, van der Grond J et al (2010) CADASIL and migraine: a narrative review. Cephalalgia 30:1284–1289

    PubMed  Google Scholar 

  31. Dichgans M, Mayer M, Uttner I et al (1998) The phenotypic spectrum of CADASIL: clinical findings in 102 cases. Ann Neurol 44:731–739

    CAS  PubMed  Google Scholar 

  32. Kaufmann P, Engelstad K, Wei Y et al (2011) Natural history of MELAS associated with mitochondrial DNA m.3243A > G genotype. Neurology 77:1965–1971

    PubMed Central  CAS  PubMed  Google Scholar 

  33. Finsterer J (2012) Inherited mitochondrial disorders. Adv Exp Med Biol 942:187–213

    CAS  PubMed  Google Scholar 

  34. Federico A, Di Donato I, Bianchi S et al (2012) Hereditary cerebral small vessel diseases: a review. J Neurol Sci 322:25–30

    PubMed  Google Scholar 

  35. Ophoff RA, DeYoung J, Service SK et al (2001) Hereditary vascular retinopathy, cerebroretinal vasculopathy, and hereditary endotheliopathy with retinopathy, nephropathy, and stroke map to a single locus on chromosome 3p21.1-p21.3. Am J Hum Genet 69:447–453

    PubMed Central  CAS  PubMed  Google Scholar 

  36. Bersano A, Debette S, Zanier ER et al (2012) The genetics of small-vessel disease. Curr Med Chem 19:4124–4141

    CAS  PubMed  Google Scholar 

  37. Storimans CW, Van Schooneveld MJ, Oosterhuis JA, Bos PJ (1991) A new autosomal dominant vascular retinopathy syndrome. Eur J Ophthalmol 1:73–78

    CAS  PubMed  Google Scholar 

  38. Terwindt GM, Haan J, Ophoff RA et al (1998) Clinical and genetic analysis of a large Dutch family with autosomal dominant vascular retinopathy, migraine and Raynaud's phenomenon. Brain 121(Pt 2):303–316

    PubMed  Google Scholar 

  39. Jen J, Cohen AH, Yue Q et al (1997) Hereditary endotheliopathy with retinopathy, nephropathy, and stroke (HERNS). Neurology 49:1322–1330

    CAS  PubMed  Google Scholar 

  40. Winkler DT, Lyrer P, Probst A et al (2008) Hereditary systemic angiopathy (HSA) with cerebral calcifications, retinopathy, progressive nephropathy, and hepatopathy. J Neurol 255:77–88

    CAS  PubMed  Google Scholar 

  41. Di Francesco JC, Novara F, Zuffardi O, et al. (2014) TREX1 C-terminal frameshift mutations in the systemic variant of retinal vasculopathy with cerebral leukodystrophy. Neurol Sci

  42. Gould DB, Phalan FC, Breedved GJ et al (2005) Mutations in COL4A1 cause perinatal cerebral hemorrhage and porencephaly. Science 308:1167–1171

    CAS  PubMed  Google Scholar 

  43. Breedved G, de Coo IF, Lequin MH et al (2006) Novel mutations in three families confirms a major role of COL4A1 in hereditary porencephaly. J Med Genet 43:490–495

    Google Scholar 

  44. Van Der Knaap MS, Smit LM, Barkhof F et al (2006) Neonatal porencephaly and adult stroke related to mutations in collagen IV A1. Ann Neurol 59:504–511

    PubMed  Google Scholar 

  45. Lanfranconi S, Markus HS (2010) COL4A1 mutations as a monogenic cause of cerebral small vessel disease: a systematic review. Stroke 41:e513–e518

    PubMed  Google Scholar 

  46. Vahedi K, Massin P, Guichard JP et al (2003) Hereditary infantile hemiparesis, retinal arteriolar tortuosity, and leukoencephalopathy. Neurology 60:57–63

    CAS  PubMed  Google Scholar 

  47. Ebisawa T (2007) Circadian rhythms in the CNS and peripheral clock disorders: human sleep disorders and clock genes. J Pharmacol Sci 103:150–154

    CAS  PubMed  Google Scholar 

  48. Vanselow K, Vanselow JT, Westermark PO et al (2006) Differential effects of PER2 phosphorylation: molecular basis for the human familial advanced sleep phase syndrome (FASPS). Genes Dev 20:2660–2672

    PubMed Central  CAS  PubMed  Google Scholar 

  49. Brennan KC, Bates EA, Shapiro RE et al (2013) Casein kinase I mutations in familial migraine and advanced sleep phase. Sci Transl Med 5(183ra56):1–11

    Google Scholar 

  50. Xu Y, Padiath QS, Shapiro RE et al (2005) Functional consequences of a CKIδ mutation causing familial advanced sleep phase syndrome. Nature 434:640–644

    CAS  PubMed  Google Scholar 

  51. Hansen JM (2010) Familial hemiplegic migraine. Dan Med Bull 57:B4183

    PubMed  Google Scholar 

  52. Thomsen LL, Kirchmann M, Bjornsson A et al (2007) The genetic spectrum of a population-based sample of familial hemiplegic migraine. Brain 130(Pt 2):346–356

    CAS  PubMed  Google Scholar 

  53. Riant F, Ducros A, Ploton C et al (2010) De novo mutations in ATP1A2 and CACNA1A are frequent in early-onset sporadic hemiplegic migraine. Neurology 75:967–972

    CAS  PubMed  Google Scholar 

  54. Ophoff RA, Terwindt GM, Vergouwe MN et al (1996) Familial hemiplegic migraine and episodic ataxia type-2 are caused by mutations in the Ca2+ channel gene CACNL1A4. Cell 87:543–552

    CAS  PubMed  Google Scholar 

  55. Ambrosini A, D’Onofrio M, Grieco G et al (2005) Familial basilar migraine associated with a new mutation in ATP1A2 gene. Neurology 65:1826–1828

    CAS  PubMed  Google Scholar 

  56. Todt U, Dichgans M, Jurkat-Rott K et al (2005) Rare missense variants in ATP1A2 in families with clustering of common forms of migraine. Hum Mutat 26:315–321

    CAS  PubMed  Google Scholar 

  57. Vanmolkot KR, Babini E, de Vries B et al (2007) The novel pL1649Q mutation in the SCN1A epilepsy gene associated with familial hemiplegic migraine: genetic and functional studies. Hum Mutat 28:522

    PubMed  Google Scholar 

  58. Cestèle S, Scalmani P, Rusconi R et al (2008) Self-limited hyperexcitability: functional effect of a familial hemiplegic migraine mutation of the Nav1.1 (SCN1A) Na+ channel. J Neurosci 28:7273–7283

    PubMed Central  PubMed  Google Scholar 

  59. Dale RC, Gardiner A, Antony J, Hulden H (2012) Familial PRRT2 mutation with heterogeneous paroxysmal disorders including paroxysmal torticollis and hemiplegic migraine. Dev Med Child Neurol 54:958–960

    PubMed  Google Scholar 

  60. Suzuki M, Van Paesschen W, Stalmans I et al (2010) Defective membrane expression of the Na(+)-HCO3(−) cotrasporter NBCe1 is associated with familial migraine. Proc Natl Acad Sci U S A 107:15963–15968

    PubMed Central  CAS  PubMed  Google Scholar 

  61. Jen JC, Wan J, Palos TP et al (2005) Mutation in the glutamate transporter EAAT1 causes episodic ataxia, hemiplegia, and seizures. Neurology 65:529–534

    CAS  PubMed  Google Scholar 

  62. de Vries B, Mamsa H, Stam AH et al (2009) Episodic ataxia associated with EAAT1 mutation C186S affecting glutamate reuptake. Arch Neurol 66:97–101

    PubMed  Google Scholar 

  63. Frosk P, Mhanni AA, Rafay MF (2013) SCN1A mutation associated with intractable myoclonic epilepsy and migraine headache. J Child Neurol 28:389–391

    PubMed  Google Scholar 

  64. Gargus JJ, Tournay A (2007) Novel mutation confirms seizure locus SCN1A is also familial hemiplegic migraine locus FHM3. Pediatr Neurol 37:407–410

    PubMed  Google Scholar 

  65. O'Roak BJ, Deriziotis P, Lee C et al (2011) Exome sequencing in sporadic autism spectrum disorders identifies severe de novo mutations. Nat Genet 43:585–589

    PubMed Central  PubMed  Google Scholar 

  66. Barros J, Ruano L, Domingos J et al (2014) The prevalence of familial hemiplegic migraine with cerebellar ataxia and spinocerebellar ataxia type 6 in Portugal. Headache 54:911–915

    PubMed  Google Scholar 

  67. Bürk K, Kaiser FJ, Tennstedt S et al (2014) A novel missense mutation in CACNA1A evaluated by in silico protein modeling is associated with non-episodic spinocerebellar ataxia with slow progression. Eur J Med Genet 57:207–211

    PubMed  Google Scholar 

  68. Star TV, Pristay W, Snutch TP (1991) Primary structure of a calcium channel that is highly expressed in the rat cerebellum. Proc Natl Acad Sci U S A 88:5621–5625

    Google Scholar 

  69. Westenbroek RE, Sakurai T, Elliot EM (1995) Immunochemical identification and subcellular distribution of the alpha 1A subunits of brain calcium channels. J Neurosci 15:6403–6418

    CAS  PubMed  Google Scholar 

  70. Cohen-Kutner M, Nachmanni D, Atlas D (2010) CaV2.1 (P/Q channel) interaction with synaptic proteins is essential for depolarization-evoked release. Channels (Austin) 4:266–277

    CAS  Google Scholar 

  71. Van Den Magdemberg AM, Pietrobon D, Pizzorusso T et al (2004) A Cacna1A knockin migraine mouse model with increased susceptibility to cortical spreading depression. Neuron 41:701–710

    Google Scholar 

  72. Eikermann-Haerter K, Yuzawa I, Qin T et al (2011) Enhanced subcortical spreading depression in familial hemiplegic migraine type 1 mutant mice. J Neurosci 31:5755–5763

    PubMed Central  CAS  PubMed  Google Scholar 

  73. Eikermann-Haerter K, Dileköz E, Kudo C 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 119:99–109

    PubMed Central  CAS  PubMed  Google Scholar 

  74. Hedrich UB, Liautard C, Kirschenbaum D et al (2014) Impaired action potential initiation in GABAergic interneurons causes hyperexcitable networks in an epileptic mouse model carrying a human NaV1.1 mutation. J Neurosci 34:14874–14889

    CAS  PubMed  Google Scholar 

  75. Escayg A, Goldin AL (2010) Sodium channel SCN1A and epilepsy: mutations and mechanisms. Epilepsia 51:1650–1658

    PubMed Central  CAS  PubMed  Google Scholar 

  76. Maher BH, Griffiths LR (2011) Identification of molecular genetic factors that influence migraine. Mol Genet Genomics 285:433–446

    CAS  PubMed  Google Scholar 

  77. Corominas R, Sobrido MJ, Ribasés M et al (2010) Association study of the serotoninergic system in migraine in the Spanish population. Am J Med Genet B Neuropsychiatr Genet 153B:177–184

    CAS  PubMed  Google Scholar 

  78. Ishii M, Shimizu S, Sakairi Y et al (2012) MAOA, MTHFR, and TNF-β genes polymorphisms and personality traits in the pathogenesis of migraine. Mol Cell Biochem 363:357–366

    CAS  PubMed  Google Scholar 

  79. Borroni B, Brambilla C, Liberini P et al (2005) Functional serotonin 5-HTTLPR polymorphism is a risk factor for migraine with aura. J Headache Pain 6:182–184

    PubMed Central  CAS  PubMed  Google Scholar 

  80. Bayerer B, Engelbergs J, Savidou I et al (2010) Single nucleotide polymorphisms of the serotonin transporter gene in migraine—an association study. Headache 50:319–322

    PubMed  Google Scholar 

  81. Lea RA, Dohy A, Jordan K et al (2000) Evidence for allelic association of the dopamine beta-hydroxylase gene (DBH) with susceptibility to typical migraine. Neurogenetics 3:35–40

    CAS  PubMed  Google Scholar 

  82. Fernandez F, Lea RA, Colson NJ, Bellis C et al (2006) Association between a 19 bp deletion polymorphism at the dopamine beta-hydroxylase (DBH) locus and migraine with aura. J Neurol Sci 251:118–123

    CAS  PubMed  Google Scholar 

  83. Fernandez F, Colson N, Quinlan S et al (2009) Association between migraine and a functional polymorphism at the dopamine beta-hydroxylase locus. Neurogenetics 10:199–208

    CAS  PubMed  Google Scholar 

  84. Todt U, Netzer C, Toliat M et al (2009) New genetic evidence for involvement of the dopamine system in migraine with aura. Hum Genet 125:265–279

    CAS  PubMed  Google Scholar 

  85. Corominas R, Ribases M, Camina M et al (2009) Two-stage case control association study of dopamine-related genes and migraine. BMC Med Genet 10:95

    PubMed Central  PubMed  Google Scholar 

  86. Ghosh J, Pradhan S, Mittal B (2011) Role of dopaminergic gene polymorphisms (DBH 19 bp indel and DRD2 Nco I) in genetic susceptibility to migraine in North Indian population. Pain Med 12:1109–1111

    PubMed  Google Scholar 

  87. Ghosh J, Pradhan S, Mittal B (2013) Identification of a novel ANKK1 and other dopaminergic (DRD2 and DBH) gene variants in migraine susceptibility. Neuromol Med 15:61–73

    CAS  Google Scholar 

  88. Mochi M, Cevoli S, Cortelli P et al (2003) A genetic association study of migraine with dopamine receptor 4, dopamine transporter and dopamine-beta-hydroxylase genes. Neurol Sci 23:301–305

    CAS  PubMed  Google Scholar 

  89. Formicola D, Aloia A, Sampaolo S et al (2010) Common variants in the regulative regions of GRIA1 and GRIA3 receptor genes are associated with migraine susceptibility. BMC Med Genet 25:103

    Google Scholar 

  90. Maher BH, Lea RA, Follett J et al (2013) Association of a GRIA3 gene polymorphism with migraine in an Australian case–control cohort. Headache 53:1245–1249

    PubMed  Google Scholar 

  91. Paterna S, Di Pasquale P, Cottone C et al (1997) Migraine without aura and ACE-gene deletion polymorphism: is there a correlation? Preliminary findings. Cardiovasc Drugs Ther 11:603–604

    CAS  PubMed  Google Scholar 

  92. Kowa H, Fusayasu E, Ijiri T et al (2005) Association of the insertion/deletion polymorphism of the angiotensin I-converting enzyme gene in patients of migraine with aura. Neurosci Lett 374:129–131

    CAS  PubMed  Google Scholar 

  93. Joshi G, Pradhan S, Mittal B (2009) Role of the ACE ID and MTHFR C677T polymorphisms in genetic susceptibility of migraine in a north Indian population. J Neurol Sci 277:133–137

    CAS  PubMed  Google Scholar 

  94. Kowa H, Yasui K, Takeshima T et al (2000) The homozygous C677T mutation in the methylenetetrahydrofolate reductase gene is a genetic risk factor for migraine. Am J Med Genet 96:762–764

    CAS  PubMed  Google Scholar 

  95. Kara I, Sazci A, Ergul E et al (2003) Association of the C677T and A1298C polymorphisms in the 5, 10 methylenetetrahydrofolate reductase gene in patients with migraine risk. Brain Res Mol Brain Res 111:84–90

    CAS  PubMed  Google Scholar 

  96. Oterino A, Valle N, Bravo Y et al (2004) MTHFR T677 homozygosis influences the presence of aura in migraineurs. Cephalalgia 24:491–494

    CAS  PubMed  Google Scholar 

  97. Lea RA, Ovcaric M, Sundholm J et al (2005) Genetic variants of angiotensin converting enzyme and methylenetetrahydrofolate reductase may act in combination to increase migraine susceptibility. Brain Res Mol Brain Res 136:112–117

    CAS  PubMed  Google Scholar 

  98. Samaan Z, Gaysina D, Cohen-Woods S et al (2011) Methylenetetrahydrofolate reductase gene variant (MTHFR C677T) and migraine: a case control study and meta-analysis. BMC Neurol 11:66

    PubMed Central  PubMed  Google Scholar 

  99. An XK, Lu CX, Ma QL et al (2013) Association of MTHFR C677T polymorphism with susceptibility to migraine in the Chinese population. Neurosci Lett 549:78–81

    CAS  PubMed  Google Scholar 

  100. Bahadir A, Eroz R, Dikici S (2013) Investigation of MTHFR C677T gene polymorphism, biochemical and clinical parameters in Turkish migraine patients: association with allodynia and fatigue. Cell Mol Neurobiol 33(8):1055–1063

    CAS  PubMed  Google Scholar 

  101. Schwaag S, Evers S, Schirmacher A et al (2006) Genetic variants of the NOTCH3 gene in migraine—a mutation analysis and association study. Cephalalgia 26:158–161

    CAS  PubMed  Google Scholar 

  102. Menon S, Cox HC, Kuwahata M et al (2010) Association of a Notch 3 gene polymorphism with migraine susceptibility. Cephalalgia 31:264–270

    PubMed  Google Scholar 

  103. Tikka-Klemola P, Kaunisto MA, Hamalainen E et al (2009) Genetic association study of endothelin-1 and its receptors EDNRA and EDNRB in migraine with aura. Cephalalgia 29:1224–1231

    Google Scholar 

  104. Joshi G, Pradhan S, Mittal B (2011) Vascular gene polymorphisms (EDNRA −231 G > A and APOE HhaI) and risk for migraine. DNA Cell Biol 30:577–584

    CAS  PubMed  Google Scholar 

  105. Lemos C, Neto JL, Pereira-Monteiro J et al (2011) A role for endothelin receptor type A in migraine without aura susceptibility? A study in Portuguese patients. Eur J Neurol 18:649–655

    CAS  PubMed  Google Scholar 

  106. Tzourio C, El Amrani M, Poirier O et al (2001) Association between migraine and endothelin type A receptor (ETA −231 A/G) gene polymorphism. Neurology 56:1273–1277

    CAS  PubMed  Google Scholar 

  107. Jia S, Ni J, Chen S et al (2011) Association of the pentanucleotide repeat polymorphism in NOS2 promoter region with susceptibility to migraine in a Chinese population. DNA Cell Biol 30:117–122

    CAS  PubMed  Google Scholar 

  108. de OS Mansur T, Goncalves FM, Martins-Oliveira A et al (2012) Inducible nitric oxide synthase haplotype associated with migraine and aura. Mol Cell Biochem 364:303–308

    Google Scholar 

  109. Borroni B, Rao R, Liberini P et al (2006) Endothelial nitric oxide synthase (Glu298Asp) polymorphism is an independent risk factor for migraine with aura. Headache 46:1575–1579

    PubMed  Google Scholar 

  110. Rainero I, Grimaldi LM, Salani G et al (2004) Association between the tumor necrosis factor-alpha −308 G/A gene polymorphism and migraine. Neurology 62:141–143

    CAS  PubMed  Google Scholar 

  111. Mazaheri S, Hajilooi M, Rafiei A (2006) The G-308A promoter variant of the tumor necrosis factor-alpha gene is associated with migraine without aura. J Neurol 253:1589–1593

    CAS  PubMed  Google Scholar 

  112. Ghosh J, Joshi G, Pradhan S, Mittal B (2010) Investigation of TNFA 308G > A and TNFB 252G > A polymorphisms in genetic susceptibility to migraine. J Neurol 257:898–904

    CAS  PubMed  Google Scholar 

  113. Yilmaz IA, Ozge A, Erdal ME et al (2010) Cytokine polymorphism in patients with migraine: some suggestive clues of migraine and inflammation. Pain Med 11:492–497

    PubMed  Google Scholar 

  114. Ates O, Kurt S, Altinisik J et al (2011) Genetic variations in tumor necrosis factor alpha, interleukin-10 genes, and migraine susceptibility. Pain Med 12:1464–1469

    PubMed  Google Scholar 

  115. Trabace S, Brioli G, Lulli P et al (2002) Tumor necrosis factor gene polymorphism in migraine. Headache 42:341–345

    PubMed  Google Scholar 

  116. Lee KA, Jang SY, Sohn KM et al (2007) Association between a polymorphism in the lymphotoxin-a promoter region and migraine. Headache 47:1056–1062

    PubMed  Google Scholar 

  117. Asuni C, Stochino ME, Cherchi A et al (2009) Migraine and tumour necrosis factor gene polymorphism. An association study in a Sardinian sample. J Neurol 256:194–197

    CAS  PubMed  Google Scholar 

  118. Dong W, Jia S, Ye X, Ni J (2012) Association analysis of TNFRSF1B polymorphism with susceptibility for migraine in the Chinese Han population. J Clin Neurosci 19:750–752

    CAS  PubMed  Google Scholar 

  119. Rainero I, Fasano E, Rubino E et al (2005) Association between migraine and HLA-DRB1 gene polymorphisms. J Headache Pain 6:185–187

    PubMed Central  CAS  PubMed  Google Scholar 

  120. Dasdemir S, Cetinkaya Y, Gencer M et al (2013) Cox-2 gene variants in migraine. Gene 518:292–295

    CAS  PubMed  Google Scholar 

  121. Lafrenière RG, Rouleau GA (2012) Identification of novel genes involved in migraine. Headache 52:107–110

    PubMed  Google Scholar 

  122. Scher AI, Eiriksdottir G, Garcia M et al (2013) Lack of association between the MTHFR C677T variant and migraine with aura in an older population: could selective survival play a role? Cephalalgia 33:308–315

    PubMed  Google Scholar 

  123. Liu R, Geng P, Ma M et al (2014) MTHFR C677T polymorphism and migraine risk: a meta-analysis. J Neurol Sci 336:68–73

    CAS  PubMed  Google Scholar 

  124. Rubino E, Ferrero M, Rainero I et al (2009) Association of the C677T polymorphism in the MTHFR gene with migraine: a meta-analysis. Cephalalgia 29:818–825

    CAS  PubMed  Google Scholar 

  125. MacClellan LR, Howard TD, Cole JW et al (2009) Relation of candidate genes that encode for endothelial function to migraine and stroke: the Stroke Prevention in Young Women study. Stroke 40:e550–e557

    PubMed Central  PubMed  Google Scholar 

  126. Pizza V, Bisogno A, Lamaida E et al (2010) Migraine and coronary artery disease: an open study on the genetic polymorphism of the 5,10 methylenetetrahydrofolate (MTHFR) and angiotensin I-converting enzyme (ACE) genes. Cent Nerv Syst Agents Med Chem 10:91–96

    CAS  PubMed  Google Scholar 

  127. Colson NJ, Lea RA, Quinlan S, Griffiths LR (2006) The role of vascular and hormonal genes in migraine susceptibility. Mol Genet Metab 88:107–113

    CAS  PubMed  Google Scholar 

  128. Stuart S, Cox HC, Lea RA, Griffiths LR (2012) The role of the MTHFR gene in migraine. Headache 52:515–520

    PubMed  Google Scholar 

  129. Schürks M, Rist PM, Kurth T (2010) MTHFR 677C > T and ACE D/I polymorphisms in migraine: a systematic review and meta-analysis. Headache 50:588–599

    PubMed Central  PubMed  Google Scholar 

  130. Colson N, Fernandez F, Griffiths L (2010) Genetics of menstrual migraine: the molecular evidence. Curr Pain Headache Rep 14:389–395

    PubMed  Google Scholar 

  131. Colson NJ, Lea RA, Quinlan S et al (2004) The estrogen receptor 1 G594A polymorphism is associated with migraine susceptibility in two independent case/control groups. Neurogenetics 5:129–133

    CAS  PubMed  Google Scholar 

  132. Oterino A, Pascual J, Ruiz de Alegria C et al (2006) Association of migraine and ESR1 G325C polymorphism. Neuroreport 17:61–64

    CAS  PubMed  Google Scholar 

  133. Oterino A, Toriello M, Cayón A et al (2008) Multilocus analyses reveal involvement of the ESR1, ESR2, and FSHR genes in migraine. Headache 48:1438–1450

    PubMed  Google Scholar 

  134. Joshi G, Pradhan S, Mittal B (2010) Role of the oestrogen receptor (ESR1 PvuII and ESR1 325 C- > G) and progesterone receptor (PROGINS) polymorphisms in genetic susceptibility to migraine in a North Indian population. Cephalalgia 30:311–320

    CAS  PubMed  Google Scholar 

  135. Ghosh J, Joshi G, Pradhan S, Mittal B (2012) Potential role of aromatase over estrogen receptor gene polymorphisms in migraine susceptibility: a case control study from North India. PLoS One 7:e34828

    PubMed Central  CAS  PubMed  Google Scholar 

  136. Colson NJ, Lea RA, Quinlan S et al (2005) Investigation of hormone receptor genes in migraine. Neurogenetics 6:17–23

    CAS  PubMed  Google Scholar 

  137. Rodriguez-Acevedo AJ, Maher BH, Lea RA et al (2013) Association of oestrogen-receptor gene (ESR1) polymorphisms with migraine in the large Norfolk Island pedigree. Cephalalgia 33:1139–1147

    PubMed  Google Scholar 

  138. 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 16:270–277

    PubMed  Google Scholar 

  139. Thompson MD, Noble-Topham S, Percy ME et al (2012) Chromosome 1p36 in migraine with aura: association study of the 5HT(1D) locus. Neuroreport 23:45–48

    CAS  PubMed  Google Scholar 

  140. Curtain RP, Lea RA, Tajouri L et al (2005) Analysis of chromosome 1 microsatellite markers and the FHM2-ATP1A2 gene mutations in migraine pedigrees. Neurol Res 27:647–652

    CAS  PubMed  Google Scholar 

  141. Nyholt DR, Morley KI, Ferreira MA et al (2005) Genome wide significant linkage to migrainous headache on chromosome 5q21. Am J Hum Genet 77:500–512

    PubMed Central  CAS  PubMed  Google Scholar 

  142. Gardner K, Barmada MM, Ptacek LJ, Hoffman EP (1997) A new locus for hemiplegic migraine maps to chromosome 1q31. Neurology 49:1231–1238

    CAS  PubMed  Google Scholar 

  143. Lea RA, Shepherd AG, Curtain RP et al (2002) A typical migraine susceptibility region localizes to chromosome 1q31. Neurogenetics 4:17–22

    CAS  PubMed  Google Scholar 

  144. Ligthart L, Nyholt DR, Hottenga JJ et al (2008) A genome-wide linkage scan provides evidence for both new and previously reported loci influencing common migraine. Am J Med Genet B Neuropsychiatr Genet 147B:1186–1195

  145. Lea RA, Nyholt DR, Curtain RP et al (2005) A genome-wide scan provides evidence for loci influencing a severe heritable form of common migraine. Neurogenetics 6:67–72

  146. Björnsson A, Gudmundsson G, Gudfinnsson E et al (2003) Localization of a gene for migraine without aura to chromosome 4q21. Am J Hum Genet 73:986–993

    PubMed Central  PubMed  Google Scholar 

  147. Wessman M, Kallela M, Kaunisto MA et al (2002) A susceptibility locus for migraine with aura, on chromosome 4q24. Am J Hum Genet 70:652–662

    PubMed Central  CAS  PubMed  Google Scholar 

  148. Anttila V, Kallela M, Oswell G et al (2006) Trait components provide tools to dissect the genetic susceptibility of migraine. Am J Hum Genet 79:85–99

    PubMed Central  CAS  PubMed  Google Scholar 

  149. Carlsson A, Forsgren L, Nylander PO et al (2002) Identification of a susceptibility locus for migraine with and without aura on 6p12.2-p21.1. Neurology 59:1804–1807

    CAS  PubMed  Google Scholar 

  150. Oterino A, Toriello M, Castillo J et al (2012) Family-based association study of chromosome 6p12.2-p21.1 migraine locus. Headache 52:393–399

    PubMed  Google Scholar 

  151. Tikka-Kleemola P, Artto V, Vepsäläinen S et al (2010) A visual migraine aura locus maps to 9q21-q22. Neurology 74:1171–1177

    PubMed Central  CAS  PubMed  Google Scholar 

  152. Anttila V, Nyholt DR, Kallela M et al (2008) Consistently replicating locus linked to migraine on 10q22-q23. Am J Hum Genet 82:1051–1063

    PubMed Central  CAS  PubMed  Google Scholar 

  153. Del Zompo M, Cherchi A, Palmas MA et al (1998) Association between dopamine receptor genes and migraine without aura in a Sardinian sample. Neurology 51:781–786

    PubMed  Google Scholar 

  154. Cader ZM, Noble-Topham S, Dyment DA et al (2003) Significant linkage to migraine with aura on chromosome 11q24. Hum Mol Genet 12:2511–2517

    CAS  PubMed  Google Scholar 

  155. Nyholt DR, Curtain RP, Gaffney PT et al (1996) Migraine association and linkage analyses of the human 5-hydroxytryptamine (5HT2A) receptor gene. Cephalalgia 16:463–467

    CAS  PubMed  Google Scholar 

  156. Soragna D, Vettori A, Carraro G et al (2003) A locus for migraine without aura maps on chromosome 14q21.2-q22.3. Am J Hum Genet 72:161–167

    PubMed Central  CAS  PubMed  Google Scholar 

  157. Russo L, Mariotti P, Sangiorgi E et al (2005) A new susceptibility locus for migraine with aura in the 15q11-q13 genomic region containing three GABA-A receptor genes. Am J Hum Genet 76:327–333

    PubMed Central  CAS  PubMed  Google Scholar 

  158. May A, Ophoff RA, Terwindt GM et al (1995) Familial hemiplegic migraine locus on 19p13 is involved in the common forms of migraine with and without aura. Hum Genet 96:604–608

    CAS  PubMed  Google Scholar 

  159. Nyholt DR, Lea RA, Goadsby PJ et al (1998) Familial typical migraine: linkage to chromosome 19p13 and evidence for genetic heterogeneity. Neurology 50:1428–1432

    CAS  PubMed  Google Scholar 

  160. Nyholt DR, Dawkins JL, Brimage PJ et al (1998) Evidence for an X-linked genetic component in familial typical migraine. Hum Mol Genet 7:459–463

    CAS  PubMed  Google Scholar 

  161. Nyholt DR, Curtain RP, Griffiths LR (2000) Familial typical migraine: significant linkage and localization of a gene to Xq24-28. Hum Genet 107:18–23

    CAS  PubMed  Google Scholar 

  162. Ducros A, Joutel A, Vahedi K et al (1997) Mapping of a second locus for familial hemiplegic migraine to 1q21-q23 and evidence of further heterogeneity. Ann Neurol 42:885–890

    CAS  PubMed  Google Scholar 

  163. Cevoli S, Pierangeli G, Monari L et al (2002) Familial hemiplegic migraine: clinical features and probable linkage to chromosome 1 in an Italian family. Neurol Sci 23:7–10

    CAS  PubMed  Google Scholar 

  164. Marconi R, De Fusco M, Aridon P et al (2003) Familial hemiplegic migraine type 2 is linked to 0.9 Mb region on chromosome 1q23. Ann Neurol 53:376–381

    CAS  PubMed  Google Scholar 

  165. Fernandez DM, Hand CK, Sweeney BJ, Parfrey NA (2008) A novel ATP1A2 gene mutation in an Irish familial hemiplegic migraine kindred. Headache 48:101–108

    PubMed  Google Scholar 

  166. Dichgans M, Freilinger T, Eckstein G et al (2005) Mutation in the neuronal voltage-gated sodium channel SCN1A in familial hemiplegic migraine. Lancet 366:371–377

    CAS  PubMed  Google Scholar 

  167. Cuenca-León E, Corominas R, Montfort M et al (2009) Familial hemiplegic migraine: linkage to chromosome 14q32 in a Spanish kindred. Neurogenetics 10:191–198

    PubMed  Google Scholar 

  168. Joutel A, Bousser MG, Biousse V et al (1994) Familial hemiplegic migraine. Localization of a responsible gene on chromosome 19. Rev Neurol (Paris) 150:340–345

    CAS  Google Scholar 

  169. Ahmed MA, Reid E, Cooke A et al (1996) Familial hemiplegic migraine in the west of Scotland: a clinical and genetic study of seven families. J Neurol Neurosurg Psychiatry 61:616–620

    PubMed Central  CAS  PubMed  Google Scholar 

  170. Lafrenière RG, Cader MZ, Poulin JF et al (2010) A dominant-negative mutation in the TRESK potassium channel is linked to familial migraine with aura. Nat Med 43:1157–1160

    Google Scholar 

  171. Lafrenière RG, Rouleau GA (2011) Role of the TRESK two-pore potassium channel. Int J Biochem Cell Biol 43:1533–1536

    PubMed  Google Scholar 

  172. Andres-Enguix I, Shang L, Stansfeld PJ et al (2012) Functional analysis of missense variants in the TRESK (KCNK18) K channel. Sci Rep 2:237

    PubMed Central  PubMed  Google Scholar 

  173. Anttila V, Stefansson H, Kallela M, International Headache Genetics Consortium et al (2010) Genome-wide association study of migraine implicates a common susceptibility variant on 8q22.1. Nat Genet 42:869–873

    PubMed Central  CAS  PubMed  Google Scholar 

  174. Kang DC, Su ZZ, Sarkar D et al (2005) Cloning and characterization of HIV-1-inducible astrocyte elevated gene-1, AEG-1. Gene 353:8–15

    CAS  PubMed  Google Scholar 

  175. Gasparini CF, Griffiths LR (2013) The biology of the glutamatergic system and potential role in migraine. Int J Biomed Sci 9:1–8

    PubMed Central  CAS  PubMed  Google Scholar 

  176. Chasman DI, Schürks M, Anttila V et al (2011) Genome-wide association study reveals three susceptibility loci for common migraine in the general population. Nat Genet 43:695–698

    PubMed Central  CAS  PubMed  Google Scholar 

  177. Proudfoot CJ, Garry EM, Cottrell DF et al (2006) Analgesia mediated by the TRPM8 cold receptor in chronic neuropathic pain. Curr Biol 16:1591–1605

    CAS  PubMed  Google Scholar 

  178. Biondi DM (2006) Is migraine a neuropathic pain syndrome? Curr Pain Headache Rep 10:167–178

    PubMed  Google Scholar 

  179. Lillis AP, Van Duyn LB, Murphy-Ullrich JE, Strickland DK (2008) LDL receptor-related protein 1: unique tissue-specific functions revealed by selective gene knockout studies. Physiol Rev 88:887–918

    PubMed Central  CAS  PubMed  Google Scholar 

  180. Arndt AK, Schafer S, Drenckhahn JD et al (2013) Fine mapping of the 1p36 deletion syndrome identifies mutation of PRDM16 as a cause of cardiomyopathy. Am J Hum Genet 93:67–77

    PubMed Central  CAS  PubMed  Google Scholar 

  181. Bjork BC, Turbe-Doan A, Prysak M et al (2010) Prdm16 is required for normal palatogenesis in mice. Hum Mol Genet 19:774–789

    PubMed Central  CAS  PubMed  Google Scholar 

  182. Fan X, Wang J, Fan W et al (2014) Replication of migraine GWAS susceptibility loci in Chinese Han population. Headache 54:709–715

    PubMed  Google Scholar 

  183. Esserlind AL, Christensen AF, Le H et al (2013) Replication and meta-analysis of common variants identifies a genome-wide significant locus in migraine. Eur J Neurol 20:765–772

    PubMed  Google Scholar 

  184. Ligthart L, de Vries B, Smith AV et al (2011) Meta-analysis of genome-wide association for migraine in six population-based European cohorts. Eur J Hum Genet 19:901–907

  185. Freilinger T, Anttila V, de Vries B, International Headache Genetics Consortium et al (2012) Genome-wide association analysis identifies susceptibility loci for migraine without aura. Nat Genet 44:777–782

    PubMed Central  CAS  PubMed  Google Scholar 

  186. Shalizi A, Gaudillière B, Yuan Z et al (2006) A calcium-regulated MEF2 sumoylation switch controls postsynaptic differentiation. Science 311:1012–1017

  187. Flavell SW, Cowan CW, Kim TK et al (2006) Activity-dependent regulation of MEF2 transcription factors suppresses excitatory synapse number. Science 311:1008–1012

    CAS  PubMed  Google Scholar 

  188. Ferrari MD, Odink J, Bos KD et al (1990) Neuroexcitatory plasma amino acids are elevated in migraine. Neurology 40:1582–1586

    CAS  PubMed  Google Scholar 

  189. Lin HY, Wang XF, Ng-Eaton E et al (1992) Expression cloning of the TGF-beta type II receptor, a functional transmembrane serine/threonine kinase. Cell 68:775–785

    CAS  PubMed  Google Scholar 

  190. Law C, Bunyan D, Castle B et al (2006) Clinical features in a family with an R460H mutation in transforming growth factor beta receptor 2 gene. J Med Genet 43:908–916

    PubMed Central  CAS  PubMed  Google Scholar 

  191. Cox HC, Lea RA, Bellis C et al (2012) A genome-wide analysis of 'Bounty’ descendants implicates several novel variants in migraine susceptibility. Neurogenetics 13:261–266

    PubMed Central  PubMed  Google Scholar 

  192. Anttila V, Winsvold BS, Gormley P, et al.; North American Brain Expression Consortium; UK Brain Expression Consortium; International Headache Genetics Consortium (2013) Genome-wide meta-analysis identifies new susceptibility loci for migraine. Nat Genet 45:912–917

  193. Russell MB, Olesen J (1995) Increased familial risk and evidence of genetic factor in migraine. BMJ 311:541–544

    PubMed Central  CAS  PubMed  Google Scholar 

  194. Persico AM, Napolioni V (2013) Autism genetics. Behav Brain Res 251:95–112

    PubMed  Google Scholar 

  195. Rudkjobing LA, Esserlind A-L, Olesen J (2012) Future possibilities in migraine genetics. J Headache Pain 13:505–511

    PubMed Central  PubMed  Google Scholar 

  196. Gottesman II, Gould TD (2003) The endophenotype concept in psychiatry: etymology and strategic intentions. Am J Psychiatry 160:636–645

    PubMed  Google Scholar 

  197. Ruggeri B, Sarkans U, Schumann G, Persico AM (2014) Biomarkers in autism spectrum disorder: the old and the new. Psychopharmacology (Berl) 231:1201–1216

    CAS  Google Scholar 

  198. Durham P, Papapetropoulos S (2013) Biomarkers associated with migraine and their potential role in migraine management. Headache 53:1262–1277

    PubMed  Google Scholar 

  199. Ligthart L, Hottenga JJ, Lewis CM et al (2014) Genetic risk score analysis indicates migraine with and without comorbid depression are genetically different disorders. Hum Genet 133:173–186

    PubMed Central  PubMed  Google Scholar 

  200. De Felice M, Ossipov MH, Wang R et al (2010) Triptan-induced enhancement of neuronal nitric oxide synthase in trigeminal ganglion dural afferents underlies increased responsiveness to potential migraine triggers. Brain 133:2475–2488

    PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgments

The authors are grateful to Drs. Maria Dora Morgante, Anna Crapulli, and to the personnel of the library at UCBM for their assistance in collecting the literature reviewed in the present article.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors. Other research by the authors is supported through the Italian Ministry for University, Scientific Research and Technology (PRIN n.2006058195 and n.2008BACT54_002), the Italian Ministry of Health (RFPS-2007-5-640174 and RF-2011-02350537), the Fondazione Gaetano e Mafalda Luce (Milan, Italy), Autism Speaks (Princeton, NJ), the Autism Research Institute (San Diego, CA), and the Innovative Medicines Initiative Joint Undertaking (EU-AIMS, n. 115300).

Conflict of interest

The authors declare that there is no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to A. M. Persico.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Persico, A.M., Verdecchia, M., Pinzone, V. et al. Migraine genetics: current findings and future lines of research. Neurogenetics 16, 77–95 (2015). https://doi.org/10.1007/s10048-014-0433-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10048-014-0433-x

Keywords

Navigation