Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Migrainomics — identifying brain and genetic markers of migraine

Key Points

  • Advances in neuroimaging and genetic studies have enabled substantial progress to be made towards the identification of migraine biomarkers

  • Brain function, structure and chemistry are altered in migraineurs versus healthy controls

  • Brain metrics such as functional MRI or voxel-based morphometry can be used as biomarkers of the disease state and treatment effects

  • Genetic findings have provided new evidence for the involvement of vascular mechanisms in migraine

  • Brain systems are also dependent on genetic determinants

  • A combination of genetic and imaging markers of migraine will deepen our understanding of migraine aetiology and improve our ability to prevent and treat attacks

Abstract

Migraine is one of the world's most prevalent and disabling disorders and imposes an enormous socioeconomic burden. The exact causes of migraine are unknown, and no recognizable diagnostic pathological changes have been identified. Specific identifiable markers of migraine would aid diagnosis and could provide insight into the pathogenesis of the condition, with the potential to direct development of new therapeutics. In the past few years, advances in neuroimaging and genetic studies have provided the most substantial progress towards the identification of markers. A growing number of brain imaging studies have provided important insights into the brain mechanisms that underlie migraine symptoms during and between migraine attacks. Similarly, large-scale genome-wide association studies have identified genetic variants associated with the common forms of migraine — migraine with aura and migraine without aura. In total, 44 independent single-nucleotide polymorphism loci have been robustly associated with the risk of migraine and provide new evidence for the involvement of vascular mechanisms. Both imaging and genetics, therefore, have excellent potential as markers of migraine. In this Review, we provide a summary of results regarding current and potential neuroimaging and genetic markers of migraine, consider what conclusions can be drawn from these markers about migraine mechanisms and discuss the potential of combining imaging and genetics.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Development of markers of migraine states.
Figure 2: Definition of MRI measures and examples of brain function and structure in migraine.
Figure 3: Word cloud of the Gene Ontology, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome pathways enriched in the 37 genes implicated in migraine.
Figure 4: Integrating imaging and genetics into markers of migraine.

Similar content being viewed by others

References

  1. GBD 2015 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet 388, 1545–1602 (2016).

  2. Vos, T. et al. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380, 2163–2196 (2012).

    PubMed  PubMed Central  Google Scholar 

  3. Linde, M. et al. The cost of headache disorders in Europe: the Eurolight project. Eur. J. Neurol. 19, 703–711 (2012).

    CAS  PubMed  Google Scholar 

  4. Launer, L. J., Terwindt, G. M. & Ferrari, M. D. The prevalence and characteristics of migraine in a population-based cohort: the GEM study. Neurology 53, 537–542 (1999).

    CAS  PubMed  Google Scholar 

  5. Saper, J. R., Silberstein, S. D., Gordon, C. D. & Hamel, R. L. Handbook of headache management: a practical guide to diagnosis and treatment of head, neck, and facial pain 1st edn (Williams & Wilkins, 1993).

    Google Scholar 

  6. Ulrich, V., Gervil, M., Kyvik, K. O., Olesen, J. & Russell, M. B. The inheritance of migraine with aura estimated by means of structural equation modelling. J. Med. Genet. 36, 225–227 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Gervil, M., Ulrich, V., Kaprio, J., Olesen, J. & Russell, M. B. The relative role of genetic and environmental factors in migraine without aura. Neurology 53, 995–999 (1999).

    CAS  PubMed  Google Scholar 

  8. Mulder, E. J. et al. Genetic and environmental influences on migraine: a twin study across six countries. Twin Res. 6, 422–431 (2003).

    PubMed  Google Scholar 

  9. Polderman, T. J. et al. Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat. Genet. 47, 702–709 (2015).

    CAS  PubMed  Google Scholar 

  10. Nyholt, D. R. & van den Maagdenberg, A. M. Genome-wide association studies in migraine: current state and route to follow. Curr. Opin. Neurol. 29, 302–308 (2016). A review of genetic association studies of migraine. Importantly, this review discusses the various strategies being tested to identify which pathophysiological mechanisms are involved, how they can be studied and what this means for clinical diagnosis and patient care.

    CAS  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  12. Coppola, G. et al. Visual evoked potentials in subgroups of migraine with aura patients. J. Headache Pain 16, 92 (2015).

    PubMed  PubMed Central  Google Scholar 

  13. Chen, W. T., Lin, Y. Y. & Wang, S. J. Headache frontiers: using magnetoencephalography to investigate pathophysiology of chronic migraine. Curr. Pain Headache Rep. 17, 309 (2013).

    PubMed  Google Scholar 

  14. Clemens, B. et al. Three-dimensional localization of abnormal EEG activity in migraine: a low resolution electromagnetic tomography (LORETA) study of migraine patients in the pain-free interval. Brain Topogr 21, 36–42 (2008).

    PubMed  Google Scholar 

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

  16. Borsook, D. et al. Sex and the migraine brain. Neurobiol. Dis. 68, 200–214 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Hubbard, C. S. et al. Brain changes in responders versus non-responders in chronic migraine: markers of disease reversal. Front. Hum. Neurosci. 10, 497 (2016).

    PubMed  PubMed Central  Google Scholar 

  18. Faria, V. et al. The migraine brain in transition: girls versus boys. Pain 156, 2212–2221 (2015). This paper was the first to look at sex differences in paediatric migraineurs during the important developmental transition between ages 10–16 years, and to examine the increases in migraine prevalence in women during and following puberty.

    PubMed  Google Scholar 

  19. Ferraro, S. et al. In medication-overuse headache, fMRI shows long-lasting dysfunction in midbrain areas. Headache 52, 1520–1534 (2012).

    PubMed  Google Scholar 

  20. Baliki, M. N., Mansour, A. R., Baria, A. T. & Apkarian, A. V. Functional reorganization of the default mode network across chronic pain conditions. PLoS ONE 9, e106133 (2014).

    PubMed  PubMed Central  Google Scholar 

  21. Borsook, D., Becerra, L. & Fava, M. Use of functional imaging across clinical phases in CNS drug development. Transl Psychiatry 3, e282 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Borsook, D., Becerra, L. & Hargreaves, R. Biomarkers for chronic pain and analgesia. Part 1: the need, reality, challenges, and solutions. Discov. Med. 11, 197–207 (2011).

    PubMed  Google Scholar 

  23. Borsook, D., Becerra, L. & Hargreaves, R. Biomarkers for chronic pain and analgesia. Part 2: how, where, and what to look for using functional imaging. Discov. Med. 11, 209–219 (2011).

    PubMed  Google Scholar 

  24. Maniyar, F. H., Sprenger, T., Monteith, T., Schankin, C. J. & Goadsby, P. J. The premonitory phase of migraine — what can we learn from it? Headache 55, 609–620 (2015).

    PubMed  Google Scholar 

  25. Schulte, L. H. & May, A. The migraine generator revisited: continuous scanning of the migraine cycle over 30 days and three spontaneous attacks. Brain 139, 1987–1993 (2016). A paper that used functional MRI to capture the migraine cycle over time and thus characterize all phases of the migraine phenotype — additional studies similar to this one are needed.

    PubMed  Google Scholar 

  26. Schulte, L. H., Allers, A. & May, A. Hypothalamus as a mediator of chronic migraine: evidence from high-resolution fMRI. Neurology 88, 2011–2016 (2017).

    PubMed  Google Scholar 

  27. Noseda, R. et al. Neural mechanism for hypothalamic-mediated autonomic responses to light during migraine. Proc. Natl Acad. Sci. USA 114, E5683–E5692 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Alstadhaug, K. B. Migraine and the hypothalamus. Cephalalgia 29, 809–817 (2009).

    CAS  PubMed  Google Scholar 

  29. Matei, D. et al. Autonomic impairment in patients with migraine. Eur. Rev. Med. Pharmacol. Sci. 19, 3922–3927 (2015).

    CAS  PubMed  Google Scholar 

  30. Denuelle, M., Fabre, N., Payoux, P., Chollet, F. & Geraud, G. Hypothalamic activation in spontaneous migraine attacks. Headache 47, 1418–1426 (2007). One of the first studies to demonstrate the importance of the hypothalamus, a key autonomic region, in migraine.

    PubMed  Google Scholar 

  31. Maniyar, F. H., Sprenger, T., Monteith, T., Schankin, C. & Goadsby, P. J. Brain activations in the premonitory phase of nitroglycerin-triggered migraine attacks. Brain 137, 232–241 (2014).

    PubMed  Google Scholar 

  32. Maniyar, F. H., Sprenger, T., Schankin, C. & Goadsby, P. J. Photic hypersensitivity in the premonitory phase of migraine — a positron emission tomography study. Eur. J. Neurol. 21, 1178–1183 (2014).

    CAS  PubMed  Google Scholar 

  33. Hadjikhani, N. et al. Mechanisms of migraine aura revealed by functional MRI in human visual cortex. Proc. Natl Acad. Sci. USA 98, 4687–4692 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Lauritzen, M. et al. Clinical relevance of cortical spreading depression in neurological disorders: migraine, malignant stroke, subarachnoid and intracranial hemorrhage, and traumatic brain injury. J. Cereb. Blood Flow Metab. 31, 17–35 (2011).

    PubMed  Google Scholar 

  35. Zhang, X. et al. Activation of meningeal nociceptors by cortical spreading depression: implications for migraine with aura. J. Neurosci. 30, 8807–8814 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Tepe, N. et al. The thalamic reticular nucleus is activated by cortical spreading depression in freely moving rats: prevention by acute valproate administration. Eur. J. Neurosci. 41, 120–128 (2015).

    PubMed  Google Scholar 

  37. Costa, C. et al. Cortical spreading depression as a target for anti-migraine agents. J. Headache Pain 14, 62 (2013).

    PubMed  PubMed Central  Google Scholar 

  38. Becerra, L. et al. Triptans disrupt brain networks and promote stress-induced CSD-like responses in cortical and subcortical areas. J. Neurophysiol. 115, 208–217 (2016).

    CAS  PubMed  Google Scholar 

  39. Hougaard, A. et al. Interhemispheric differences of fMRI responses to visual stimuli in patients with side-fixed migraine aura. Hum. Brain Mapp. 35, 2714–2723 (2014).

    PubMed  Google Scholar 

  40. Datta, R., Aguirre, G. K., Hu, S., Detre, J. A. & Cucchiara, B. Interictal cortical hyperresponsiveness in migraine is directly related to the presence of aura. Cephalalgia 33, 365–374 (2013).

    PubMed  PubMed Central  Google Scholar 

  41. Borsook, D., DaSilva, A. F., Ploghaus, A. & Becerra, L. Specific and somatotopic functional magnetic resonance imaging activation in the trigeminal ganglion by brush and noxious heat. J. Neurosci. 23, 7897–7903 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Stankewitz, A., Aderjan, D., Eippert, F. & May, A. Trigeminal nociceptive transmission in migraineurs predicts migraine attacks. J. Neurosci. 31, 1937–1943 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Borsook, D. & Burstein, R. The enigma of the dorsolateral pons as a migraine generator. Cephalalgia 32, 803–812 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Burstein, R. et al. Thalamic sensitization transforms localized pain into widespread allodynia. Ann. Neurol. 68, 81–91 (2010).

    PubMed  PubMed Central  Google Scholar 

  45. DaSilva, A. F. et al. 3D-neuronavigation in vivo through a patient's brain during a spontaneous migraine headache. J. Vis. Exp. http://dx.doi.org/10.3791/50682 (2014).

  46. Moulton, E. A. et al. Painful heat reveals hyperexcitability of the temporal pole in interictal and ictal migraine states. Cereb. Cortex 21, 435–448 (2011).

    CAS  PubMed  Google Scholar 

  47. Stankewitz, A. & May, A. Increased limbic and brainstem activity during migraine attacks following olfactory stimulation. Neurology 77, 476–482 (2011).

    PubMed  Google Scholar 

  48. DaSilva, A. F. et al. Association of μ-opioid activation in the prefrontal cortex with spontaneous migraine attacks — brief report I. Ann. Clin. Transl Neurol. 1, 439–444 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Akerman, S., Holland, P. R. & Goadsby, P. J. Diencephalic and brainstem mechanisms in migraine. Nat. Rev. Neurosci. 12, 570–584 (2011).

    CAS  PubMed  Google Scholar 

  50. Bahra, A., Matharu, M. S., Buchel, C., Frackowiak, R. S. & Goadsby, P. J. Brainstem activation specific to migraine headache. Lancet 357, 1016–1017 (2001).

    CAS  PubMed  Google Scholar 

  51. Beissner, F. & Baudrexel, S. Investigating the human brainstem with structural and functional MRI. Front. Hum. Neurosci. 8, 116 (2014).

    PubMed  PubMed Central  Google Scholar 

  52. Henderson, L. A. & Macefield, V. G. Functional imaging of the human brainstem during somatosensory input and autonomic output. Front. Hum. Neurosci. 7, 569 (2013).

    PubMed  PubMed Central  Google Scholar 

  53. Moulton, E. A., Becerra, L., Johnson, A., Burstein, R. & Borsook, D. Altered hypothalamic functional connectivity with autonomic circuits and the locus coeruleus in migraine. PLoS ONE 9, e95508 (2014).

    PubMed  PubMed Central  Google Scholar 

  54. Mainero, C., Boshyan, J. & Hadjikhani, N. Altered functional magnetic resonance imaging resting-state connectivity in periaqueductal gray networks in migraine. Ann. Neurol. 70, 838–845 (2011). An example of specific circuits involved in brainstem modulation in migraine.

    PubMed  PubMed Central  Google Scholar 

  55. Mulder, E. J., Linssen, W. H., Passchier, J., Orlebeke, J. F. & de Geus, E. J. Interictal and postictal cognitive changes in migraine. Cephalalgia 19, 557–565; discussion 541 (1999).

    CAS  PubMed  Google Scholar 

  56. Bramanti, P. et al. Ictal and interictal hypoactivation of the occipital cortex in migraine with aura. A neuroimaging and electrophysiological study. Funct. Neurol. 20, 169–171 (2005).

    PubMed  Google Scholar 

  57. Maleki, N. et al. Concurrent functional and structural cortical alterations in migraine. Cephalalgia 32, 607–620 (2012).

    PubMed  Google Scholar 

  58. Grazzi, L. et al. Chronic migraine with medication overuse pre-post withdrawal of symptomatic medication: clinical results and fMRI correlations. Headache 50, 998–1004 (2010).

    PubMed  Google Scholar 

  59. Maleki, N. et al. Her versus his migraine: multiple sex differences in brain function and structure. Brain 135, 2546–2559 (2012). Sex differences in migraine are well defined; this paper reports significant differences between men and women that could contribute to how brain markers of disease need to be dimorphically segregated.

    PubMed  PubMed Central  Google Scholar 

  60. Mehnert, J. & May, A. Functional and structural alterations in the migraine cerebellum. J. Cereb. Blood Flow Metab. http://dx.doi.org/10.1177/0271678X17722109 (2017).

  61. Maleki, N. et al. Migraine attacks the basal ganglia. Mol. Pain 7, 71 (2011).

    PubMed  PubMed Central  Google Scholar 

  62. Rocca, M. A. et al. Structural brain MRI abnormalities in pediatric patients with migraine. J. Neurol. 261, 350–357 (2014).

    PubMed  Google Scholar 

  63. Riederer, F. et al. Decrease of gray matter volume in the midbrain is associated with treatment response in medication-overuse headache: possible influence of orbitofrontal cortex. J. Neurosci. 33, 15343–15349 (2013). Migraine is a dynamic state, and biomarker evaluation must take this fact into account; this paper examines migraine responsivity (that is, brain changes) in responders versus non-responders to reversal of medication overuse.

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Borsook, D., Erpelding, N. & Becerra, L. Losses and gains: chronic pain and altered brain morphology. Expert Rev. Neurother. 13, 1221–1234 (2013).

    CAS  PubMed  Google Scholar 

  65. Hu, W., Guo, J., Chen, N., Guo, J. & He, L. A meta-analysis of voxel-based morphometric studies on migraine. Int. J. Clin. Exp. Med. 8, 4311–4319 (2015).

    PubMed  PubMed Central  Google Scholar 

  66. Hougaard, A., Amin, F. M. & Ashina, M. Migraine and structural abnormalities in the brain. Curr. Opin. Neurol. 27, 309–314 (2014).

    PubMed  Google Scholar 

  67. Kruit, M. C., van Buchem, M. A., Launer, L. J., Terwindt, G. M. & Ferrari, M. D. Migraine is associated with an increased risk of deep white matter lesions, subclinical posterior circulation infarcts and brain iron accumulation: the population-based MRI CAMERA study. Cephalalgia 30, 129–136 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Aguila, M. R. et al. The association between clinical characteristics of migraine and brain GABA levels: an exploratory study. J. Pain 17, 1058–1067 (2016).

    CAS  PubMed  Google Scholar 

  69. Aguila, M. E. et al. Elevated levels of GABA+ in migraine detected using 1H-MRS. NMR Biomed. 28, 890–897 (2015). An understanding of the chemical changes in the brains of migraineurs, such as those examined in this study, will become increasingly important, as these alterations present potential biomarkers and targets for therapy development.

    CAS  PubMed  Google Scholar 

  70. Riederer, F. et al. Grey matter changes associated with medication-overuse headache: correlations with disease related disability and anxiety. World J. Biol. Psychiatry 13, 517–525 (2012).

    PubMed  Google Scholar 

  71. Kim, J. H. et al. Thickening of the somatosensory cortex in migraine without aura. Cephalalgia 34, 1125–1133 (2014).

    PubMed  Google Scholar 

  72. DaSilva, A. F., Granziera, C., Snyder, J. & Hadjikhani, N. Thickening in the somatosensory cortex of patients with migraine. Neurology 69, 1990–1995 (2007).

    PubMed  Google Scholar 

  73. Abanoz, Y., Abanoz, Y., Gunduz, A. & Savrun, F. K. Trigeminal somatosensorial evoked potentials suggest increased excitability during interictal period in patients with long disease duration in migraine. Neurosci. Lett. 612, 62–65 (2016).

    CAS  PubMed  Google Scholar 

  74. Hodkinson, D. J. et al. Primary somatosensory cortices contain altered patterns of regional cerebral blood flow in the interictal phase of migraine. PLoS ONE 10, e0137971 (2015).

    PubMed  PubMed Central  Google Scholar 

  75. Pierelli, F., Iacovelli, E., Bracaglia, M., Serrao, M. & Coppola, G. Abnormal sensorimotor plasticity in migraine without aura patients. Pain 154, 1738–1742 (2013).

    PubMed  Google Scholar 

  76. Yuan, K. et al. Altered structure and resting-state functional connectivity of the basal ganglia in migraine patients without aura. J. Pain 14, 836–844 (2013).

    PubMed  Google Scholar 

  77. Schwedt, T. J. et al. Atypical resting-state functional connectivity of affective pain regions in chronic migraine. Headache 53, 737–751 (2013). Evaluation of resting states in the brain, as was conducted in this study, could be important for understanding disease state and treatment effects.

    PubMed  PubMed Central  Google Scholar 

  78. Tso, A. R., Trujillo, A., Guo, C. C., Goadsby, P. J. & Seeley, W. W. The anterior insula shows heightened interictal intrinsic connectivity in migraine without aura. Neurology 84, 1043–1050 (2015).

    PubMed  PubMed Central  Google Scholar 

  79. Hadjikhani, N. et al. The missing link: enhanced functional connectivity between amygdala and visceroceptive cortex in migraine. Cephalalgia 33, 1264–1268 (2013).

    PubMed  PubMed Central  Google Scholar 

  80. Wilcox, S. L. et al. Increased functional activation of limbic brain regions during negative emotional processing in migraine. Front. Hum. Neurosci. 10, 366 (2016). This paper was important in demonstrating that brain systems involved in emotional processing are altered in migraineurs, even in the interictal state.

    PubMed  PubMed Central  Google Scholar 

  81. Loggia, M. L. et al. The lateral prefrontal cortex mediates the hyperalgesic effects of negative cognitions in chronic pain patients. J. Pain 16, 692–699 (2015).

    PubMed  PubMed Central  Google Scholar 

  82. Hashmi, J. A. et al. Shape shifting pain: chronification of back pain shifts brain representation from nociceptive to emotional circuits. Brain 136, 2751–2768 (2013).

    PubMed  PubMed Central  Google Scholar 

  83. Prescot, A. et al. Excitatory neurotransmitters in brain regions in interictal migraine patients. Mol. Pain 5, 34 (2009).

    PubMed  PubMed Central  Google Scholar 

  84. Bridge, H. et al. Altered neurochemical coupling in the occipital cortex in migraine with visual aura. Cephalalgia 35, 1025–1030 (2015).

    PubMed  Google Scholar 

  85. Harris, R. E. et al. Dynamic levels of glutamate within the insula are associated with improvements in multiple pain domains in fibromyalgia. Arthritis Rheum. 58, 903–907 (2008).

    CAS  PubMed  Google Scholar 

  86. Schwedt, T. J. et al. Allodynia and descending pain modulation in migraine: a resting state functional connectivity analysis. Pain Med. 15, 154–165 (2014).

    PubMed  Google Scholar 

  87. Moulton, E. A. et al. Interictal dysfunction of a brainstem descending modulatory center in migraine patients. PLoS ONE 3, e3799 (2008).

    PubMed  PubMed Central  Google Scholar 

  88. Li, Z. et al. Altered periaqueductal gray resting state functional connectivity in migraine and the modulation effect of treatment. Sci. Rep. 6, 20298 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Borsook, D., Maleki, N., Becerra, L. & McEwen, B. Understanding migraine through the lens of maladaptive stress responses: a model disease of allostatic load. Neuron 73, 219–234 (2012). A summary of the complexity of processes that could affect the migraine brain in a dynamic manner, which need to be taken into account in brain biomarker development.

    CAS  PubMed  Google Scholar 

  90. Bigal, M. E. et al. Acute migraine medications and evolution from episodic to chronic migraine: a longitudinal population-based study. Headache 48, 1157–1168 (2008).

    PubMed  Google Scholar 

  91. Bigal, M. E. & Lipton, R. B. Concepts and mechanisms of migraine chronification. Headache 48, 7–15 (2008).

    PubMed  Google Scholar 

  92. Bigal, M. E. & Lipton, R. B. Migraine chronification. Curr. Neurol. Neurosci. Rep. 11, 139–148 (2011).

    PubMed  Google Scholar 

  93. Aguggia, M. & Saracco, M. G. Pathophysiology of migraine chronification. Neurol. Sci. 31, S15–S17 (2010).

    PubMed  Google Scholar 

  94. Bigal, M. E. & Lipton, R. B. Overuse of acute migraine medications and migraine chronification. Curr. Pain Headache Rep. 13, 301–307 (2009).

    PubMed  Google Scholar 

  95. Chiapparini, L., Ferraro, S., Grazzi, L. & Bussone, G. Neuroimaging in chronic migraine. Neurol. Sci. 31, S19–S22 (2010).

    PubMed  Google Scholar 

  96. Chiapparini, L. et al. Functional-MRI evaluation of pain processing in chronic migraine with medication overuse. Neurol. Sci. 30, S71–S74 (2009). Understanding the effects of drugs on the migraine brain is key to the evaluation of biomarkers; the natural history of migraine chronification resulting from medication overuse, as studied in this paper, is a good model with which to achieve this understanding.

    PubMed  Google Scholar 

  97. Aurora, S. K., Barrodale, P. M., Tipton, R. L. & Khodavirdi, A. Brainstem dysfunction in chronic migraine as evidenced by neurophysiological and positron emission tomography studies. Headache 47, 996–1003; discussion 1004–1007 (2007).

    PubMed  Google Scholar 

  98. Lai, T. H. et al. Gray matter changes related to medication overuse in patients with chronic migraine. Cephalalgia 36, 1324–1333 (2016).

    PubMed  Google Scholar 

  99. Finocchi, C. & Strada, L. Sex-related differences in migraine. Neurol. Sci. 35 (Suppl. 1), 207–213 (2014).

    PubMed  Google Scholar 

  100. Borsook, D., Hargreaves, R. & Becerra, L. Can functional magnetic resonance imaging improve success rates in CNS drug discovery? Expert Opin. Drug Discov. 6, 597–617 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Kroger, I. L. & May, A. Triptan-induced disruption of trigemino-cortical connectivity. Neurology 84, 2124–2131 (2015).

    PubMed  Google Scholar 

  102. Borsook, D. et al. The insula: a “hub of activity” in migraine. Neuroscientist 22, 632–652 (2015).

    PubMed  PubMed Central  Google Scholar 

  103. Ophoff, R. A. et al. Familial hemiplegic migraine and episodic ataxia type-2 are caused by mutations in the Ca2+ channel gene CACNL1A4. Cell 87, 543–552 (1996).

    CAS  PubMed  Google Scholar 

  104. De Fusco, M. et al. Haploinsufficiency of ATP1A2 encoding the Na+/K+ pump α2 subunit associated with familial hemiplegic migraine type 2. Nat. Genet. 33, 192–196 (2003).

    CAS  PubMed  Google Scholar 

  105. Dichgans, M. et al. Mutation in the neuronal voltage-gated sodium channel SCN1A in familial hemiplegic migraine. Lancet 366, 371–377 (2005).

    CAS  PubMed  Google Scholar 

  106. Garza-Lopez, E. et al. Familial hemiplegic migraine type 1 mutations W1684R and V1696I alter G protein-mediated regulation of CaV2.1 voltage-gated calcium channels. Biochim. Biophys. Acta 1822, 1238–1246 (2012).

    CAS  PubMed  Google Scholar 

  107. Thomsen, L. L. et al. A population-based study of familial hemiplegic migraine suggests revised diagnostic criteria. Brain 125, 1379–1391 (2002).

    CAS  PubMed  Google Scholar 

  108. Lafreniere, R. G. et al. A dominant-negative mutation in the TRESK potassium channel is linked to familial migraine with aura. Nat. Med. 16, 1157–1160 (2010).

    CAS  PubMed  Google Scholar 

  109. Brennan, K. C. et al. Casein kinase iδ mutations in familial migraine and advanced sleep phase. Sci. Transl. Med. 5, 183ra56 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Hirschhorn, J. N., Lohmueller, K., Byrne, E. & Hirschhorn, K. A comprehensive review of genetic association studies. Genet. Med. 4, 45–61 (2002).

    CAS  PubMed  Google Scholar 

  111. Button, K. S. et al. Power failure: why small sample size undermines the reliability of neuroscience. Nat. Rev. Neurosci. 14, 365–376 (2013).

    CAS  PubMed  Google Scholar 

  112. de Vries, B. et al. Systematic re-evaluation of genes from candidate gene association studies in migraine using a large genome-wide association data set. Cephalalgia 36, 604–614 (2016). This comprehensive review of 27 genes from published candidate gene and non-genome-wide association studies in migraine finds no clear evidence for involvement of the previously reported most promising candidate genes in migraine.

    PubMed  Google Scholar 

  113. Nyholt, D. R. et al. A high-density association screen of 155 ion transport genes for involvement with common migraine. Hum. Mol. Genet. 17, 3318–3331 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. McCarthy, M. I. et al. Genome-wide association studies for complex traits: consensus, uncertainty and challenges. Nat. Rev. Genet. 9, 356–369 (2008).

    CAS  PubMed  Google Scholar 

  115. Anttila, V. et al. Genome-wide association study of migraine implicates a common susceptibility variant on 8q22.1. Nat. Genet. 42, 869–873 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Freilinger, T. et al. Genome-wide association analysis identifies susceptibility loci for migraine without aura. Nat. Genet. 44, 777–782 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Chasman, D. I. et al. Genome-wide association study reveals three susceptibility loci for common migraine in the general population. Nat. Genet. 43, 695–698 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Anttila, V. et al. Genome-wide meta-analysis identifies new susceptibility loci for migraine. Nat. Genet. 45, 912–917 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Nyholt, D. R. SECA: SNP effect concordance analysis using genome-wide association summary results. Bioinformatics 30, 2086–2088 (2014).

    CAS  PubMed  Google Scholar 

  121. Nyholt, D. R. et al. Concordance of genetic risk across migraine subgroups: impact on current and future genetic association studies. Cephalalgia 35, 489–499 (2015). This thorough analysis of genome-wide association results from Ref. 119 shows that the majority of common genetic risk effects are the same across migraine without aura and migraine with aura subgroups, clinic-based and population-based subgroups and male and female patients with migraine subgroups.

    PubMed  Google Scholar 

  122. Visscher, P. M., Brown, M. A., McCarthy, M. I. & Yang, J. Five years of GWAS discovery. Am. J. Hum. Genet. 90, 7–24 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Gormley, P. et al. Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat. Genet. 48, 856–866 (2016). This is the largest genetic study of migraine to date; the identified loci showed enrichment of genes expressed in vascular and smooth muscle tissues, consistent with a predominant theory of migraine that highlights vascular aetiologies.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Chasman, D. I. et al. Selectivity in genetic association with sub-classified migraine in women. PLoS Genet. 10, e1004366 (2014).

    PubMed  PubMed Central  Google Scholar 

  125. 1000 Genomes Project Consortium. et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

  126. Wellcome Trust Case Control Consortium et al. Bayesian refinement of association signals for 14 loci in 3 common diseases. Nat. Genet. 44, 1294–1301 (2012).

  127. Pers, T. H. et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nat. Commun. 6, 5890 (2015).

    CAS  PubMed  Google Scholar 

  128. Reimand, J. et al. g:Profiler — a web server for functional interpretation of gene lists (2016 update). Nucleic Acids Res. 44, W83–W89 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Ashburner, M. et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 25, 25–29 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Kanehisa, M. & Goto, S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28, 27–30 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Kanehisa, M., Sato, Y., Kawashima, M., Furumichi, M. & Tanabe, M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 44, 457–D462 (2016).

    Google Scholar 

  132. Milacic, M. et al. Annotating cancer variants and anti-cancer therapeutics in reactome. Cancers (Basel) 4, 1180–1211 (2012).

    CAS  Google Scholar 

  133. Croft, D. et al. The reactome pathway knowledgebase. Nucleic Acids Res. 42, D472–D477 (2014).

    CAS  PubMed  Google Scholar 

  134. GTEx Consortium. Human genomics. The genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

  135. Malik, R. et al. Shared genetic basis for migraine and ischemic stroke: a genome-wide analysis of common variants. Neurology 84, 2132–2145 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Winsvold, B. S. et al. Genetic analysis for a shared biological basis between migraine and coronary artery disease. Neurol. Genet. 1, e10 (2015).

    PubMed  PubMed Central  Google Scholar 

  137. Debette, S. et al. Common variation in PHACTR1 is associated with susceptibility to cervical artery dissection. Nat. Genet. 47, 78–83 (2015).

    CAS  PubMed  Google Scholar 

  138. Myocardial Infarction Genetics Consortium. et al. Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat. Genet. 41, 334–341 (2009).

  139. Law, C. et al. Clinical features in a family with an R460H mutation in transforming growth factor β receptor 2 gene. J. Med. Genet. 43, 908–916 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Bown, M. J. et al. Abdominal aortic aneurysm is associated with a variant in low-density lipoprotein receptor-related protein 1. Am. J. Hum. Genet. 89, 619–627 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Arndt, A. K. et al. Fine mapping of the 1p36 deletion syndrome identifies mutation of PRDM16 as a cause of cardiomyopathy. Am. J. Hum. Genet. 93, 67–77 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Fujimura, M. et al. Genetics and biomarkers of moyamoya disease: significance of RNF213 as a susceptibility gene. J. Stroke 16, 65–72 (2014).

    PubMed  PubMed Central  Google Scholar 

  143. McElhinney, D. B. et al. Analysis of cardiovascular phenotype and genotype-phenotype correlation in individuals with a JAG1 mutation and/or Alagille syndrome. Circulation 106, 2567–2574 (2002).

    PubMed  Google Scholar 

  144. Bezzina, C. R. et al. Common variants at SCN5A-SCN10A and HEY2 are associated with Brugada syndrome, a rare disease with high risk of sudden cardiac death. Nat. Genet. 45, 1044–1049 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Sinner, M. F. et al. Integrating genetic, transcriptional, and functional analyses to identify 5 novel genes for atrial fibrillation. Circulation 130, 1225–1235 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Neale, B. M. et al. Genome-wide association study of advanced age-related macular degeneration identifies a role of the hepatic lipase gene (LIPC). Proc. Natl Acad. Sci. USA 107, 7395–7400 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Zach, V., Bezov, D., Lipton, R. B. & Ashina, S. Headache associated with moyamoya disease: a case story and literature review. J. Headache Pain 11, 79–82 (2010).

    PubMed  Google Scholar 

  148. Porcu, E. et al. A meta-analysis of thyroid-related traits reveals novel loci and gender-specific differences in the regulation of thyroid function. PLoS Genet. 9, e1003266 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Fenichel, N. M. Chronic headache due to masked hypothyroidism. Ann. Intern. Med. 29, 456–460 (1948).

    CAS  PubMed  Google Scholar 

  150. Bigal, M. E., Sheftell, F. D., Rapoport, A. M., Tepper, S. J. & Lipton, R. B. Chronic daily headache: identification of factors associated with induction and transformation. Headache 42, 575–581 (2002).

    PubMed  Google Scholar 

  151. Martin, A. T. et al. Headache disorders may be a risk factor for the development of new onset hypothyroidism. Headache 57, 21–30 (2017).

    PubMed  Google Scholar 

  152. Mehta, N. N. A genome-wide association study in Europeans and South Asians identifies 5 new loci for coronary artery disease. Circ. Cardiovasc. Genet. 4, 465–466 (2011).

    PubMed  PubMed Central  Google Scholar 

  153. O'Donnell, C. J. et al. Genome-wide association study for coronary artery calcification with follow-up in myocardial infarction. Circulation 124, 2855–2864 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Dichgans, M. et al. Shared genetic susceptibility to ischemic stroke and coronary artery disease: a genome-wide analysis of common variants. Stroke 45, 24–36 (2014).

    CAS  PubMed  Google Scholar 

  155. Soler Artigas, M. et al. Genome-wide association and large-scale follow up identifies 16 new loci influencing lung function. Nat. Genet. 43, 1082–1090 (2011).

    PubMed  Google Scholar 

  156. Ripke, S. et al. Genome-wide association analysis identifies 13 new risk loci for schizophrenia. Nat. Genet. 45, 1150–1159 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Yu, B. et al. Genome-wide association study of a heart failure related metabolomic profile among African Americans in the atherosclerosis risk in communities (ARIC) study. Genet. Epidemiol. 37, 840–845 (2013).

    PubMed  PubMed Central  Google Scholar 

  158. Imboden, M. et al. Genome-wide association study of lung function decline in adults with and without asthma. J. Allergy Clin. Immunol. 129, 1218–1228 (2012).

    PubMed  PubMed Central  Google Scholar 

  159. He, J. et al. Genome-wide association study identifies 8 novel loci associated with blood pressure responses to interventions in Han Chinese. Circ. Cardiovasc. Genet. 6, 598–607 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Johnson, A. D. et al. Genome-wide meta-analyses identifies seven loci associated with platelet aggregation in response to agonists. Nat. Genet. 42, 608–613 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Schunkert, H. et al. Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat. Genet. 43, 333–338 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Bis, J. C. et al. Common variants at 12q14 and 12q24 are associated with hippocampal volume. Nat. Genet. 44, 545–551 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Gasser, U. E. & Hatten, M. E. Neuron-glia interactions of rat hippocampal cells in vitro: glial-guided neuronal migration and neuronal regulation of glial differentiation. J. Neurosci. 10, 1276–1285 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Wilson, P. M., Fryer, R. H., Fang, Y. & Hatten, M. E. Astn2, a novel member of the astrotactin gene family, regulates the trafficking of ASTN1 during glial-guided neuronal migration. J. Neurosci. 30, 8529–8540 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Wagner, J. A. Strategic approach to fit-for-purpose biomarkers in drug development. Annu. Rev. Pharmacol. Toxicol. 48, 631–651 (2008).

    CAS  PubMed  Google Scholar 

  166. Oberndorfer, S. et al. Familial hemiplegic migraine: follow-up findings of diffusion-weighted magnetic resonance imaging (MRI), perfusion-MRI and [99mTc] HMPAO-SPECT in a patient with prolonged hemiplegic aura. Cephalalgia 24, 533–539 (2004).

    CAS  PubMed  Google Scholar 

  167. Lindahl, A. J. et al. Prolonged hemiplegic migraine associated with unilateral hyperperfusion on perfusion weighted magnetic resonance imaging. J. Neurol. Neurosurg. Psychiatry 73, 202–203 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Yang, Y. et al. Genetic epidemiology of migraine and depression. Cephalalgia 36, 679–691 (2016).

    PubMed  Google Scholar 

  169. Marx, V. Biology: the big challenges of big data. Nature 498, 255–260 (2013).

    CAS  PubMed  Google Scholar 

  170. Van Horn, J. D. & Toga, A. W. Human neuroimaging as a “big data” science. Brain Imaging Behav. 8, 323–331 (2014).

    PubMed  PubMed Central  Google Scholar 

  171. Turk-Browne, N. B. Functional interactions as big data in the human brain. Science 342, 580–584 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Consortium of the Human Genome Project. The NIH human connectome project. University of Southern California http://www.humanconnectomeproject.org (2016).

  173. Thompson, P. M., Martin, N. G. & Wright, M. J. Imaging genomics. Curr. Opin. Neurol. 23, 368–373 (2010).

    PubMed  PubMed Central  Google Scholar 

  174. ENIGMA. Enhancing neuro imaging genetics through meta analysis. University of Southern California http://enigma.ini.usc.edu (2016).

  175. Thompson, P. M. et al. ENIGMA and the individual: predicting factors that affect the brain in 35 countries worldwide. Neuroimage 145, 389–408 (2015).

    PubMed  Google Scholar 

  176. Levy, M., Lu, Z., Dion, G. & Kara, P. The shape of dendritic arbors in different functional domains of the cortical orientation map. J. Neurosci. 34, 3231–3236 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Atluri, V. S., Kanthikeel, S. P., Reddy, P. V., Yndart, A. & Nair, M. P. Human synaptic plasticity gene expression profile and dendritic spine density changes in HIV-infected human CNS cells: role in HIV-associated neurocognitive disorders (HAND). PLoS ONE 8, e61399 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Pickrell, J. K. et al. Detection and interpretation of shared genetic influences on 42 human traits. Nat. Genet. 48, 709–717 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Zielman, R. et al. Biochemical changes in the brain of hemiplegic migraine patients measured with 7 tesla 1H-MRS. Cephalalgia 34, 959–967 (2014).

    CAS  PubMed  Google Scholar 

  180. Blicher, J. U., Tietze, A., Donahue, M. J., Smith, S. A. & Ostergaard, L. Perfusion and pH MRI in familial hemiplegic migraine with prolonged aura. Cephalalgia 36, 279–283 (2016).

    PubMed  Google Scholar 

  181. Kim, S., Kang, M. & Choi, S. A case report of sporadic hemiplegic migraine associated cerebral hypoperfusion: comparison of arterial spin labeling and dynamic susceptibility contrast perfusion MR imaging. Eur. J. Pediatr. 175, 295–298 (2016).

    PubMed  Google Scholar 

  182. Cutrer, F. M. & Smith, J. H. Human studies in the pathophysiology of migraine: genetics and functional neuroimaging. Headache 53, 401–412 (2013).

    PubMed  Google Scholar 

  183. Li, Z. et al. The altered right frontoparietal network functional connectivity in migraine and the modulation effect of treatment. Cephalalgia 37, 161–176 (2017).

    PubMed  Google Scholar 

  184. Messina, R. et al. White matter microstructure abnormalities in pediatric migraine patients. Cephalalgia 35, 1278–1286 (2015).

    PubMed  Google Scholar 

  185. Becerra, L. et al. A 'complex' of brain metabolites distinguish altered chemistry in the cingulate cortex of episodic migraine patients. Neuroimage Clin. 11, 588–594 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. R Development Core Team. The R project for statistical computing. The R Foundation http://www.R-project.org/ (2017).

Download references

Acknowledgements

D.R.N. is supported in part by a National Health and Medical Research Council (NHMRC) project grant (APP1075175) and the European Union Seventh Framework Programme (2007–2013) under grant agreement no. 602633 (EUROHEADPAIN). D.B. is supported by grants from the NIH (NINDS Grants: K24NS064050, R01NS0750182, RO1 NS073977) and by the Mayday/Louis Herlands Chair for Pain Systems Science and the National Headache Foundation. L.R.G.'s migraine research is supported by NHMRC project grants APP1058808 and APP1083450.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed equally to all stages of the preparation of the manuscript.

Corresponding authors

Correspondence to Dale R. Nyholt, David Borsook or Lyn R. Griffiths.

Ethics declarations

Competing interests

D.R.N. declares no competing financial interests. L.R.G. consults for Novartis and D.B. consults for Biogen.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nyholt, D., Borsook, D. & Griffiths, L. Migrainomics — identifying brain and genetic markers of migraine. Nat Rev Neurol 13, 725–741 (2017). https://doi.org/10.1038/nrneurol.2017.151

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneurol.2017.151

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research