Skip to main content
Top
Published in: The Egyptian Journal of Neurology, Psychiatry and Neurosurgery 1/2019

Open Access 01-12-2019 | Migraine | Review

Role of single nucleotide polymorphisms (SNPs) in common migraine

Authors: Sukhvinder Kaur, Arif Ali, Uzair Ahmad, Yaser Siahbalaei, A. K. Pandey, Balkirat Singh

Published in: The Egyptian Journal of Neurology, Psychiatry and Neurosurgery | Issue 1/2019

Login to get access

Abstract

Single nucleotide polymorphisms (SNPs) may act as biological markers, as they can relate to the genes that are associated with various complex diseases such as heart diseases, diabetes, cancer, schizophrenia, blood pressure, migraine, and Alzheimer. These SNPs mostly locate within a gene or in a regulatory region near a gene and can affect the gene’s function to play a more direct role in disease. Hence, SNPs allow scientists to develop candidate drug therapy by evaluating an individual’s genetic makeup to develop a particular disease. Gene-gene interactions generally complicate migraine and its genetics and further gene-environmental interactions that often misguide the true defying causes of this disease. Due to its complex nature, it is difficult for scientists to reveal a complete list of SNPs or even all the genes that are related with the pathogenesis of this disease. Nowadays, much work has been done in this direction and new variants of migraine are being constantly identified. In this review article, the role of various SNPs reported to be disease-associated in published migraine GWAS has been discussed. To understand the molecular mechanisms of migraine attack by identifying new genetic variants of migraine can be a key to develop new therapeutic strategies in the future.
Literature
2.
go back to reference Stewart WF, Shechter A, Rasmussen BK. Migraine prevalence: a review of population-based studies. Neurology. 1994;44(6 Suppl 4):S17–23.PubMed Stewart WF, Shechter A, Rasmussen BK. Migraine prevalence: a review of population-based studies. Neurology. 1994;44(6 Suppl 4):S17–23.PubMed
4.
go back to reference Adams AM, Serrano D, Buse DC, Reed ML, Marske V, et al. The impact of chronic migraine: the Chronic Migraine Epidemiology and Outcomes (CaMEO) Study methods and baseline results. Cephalalgia. 2015;35(7):563–78.PubMedPubMedCentralCrossRef Adams AM, Serrano D, Buse DC, Reed ML, Marske V, et al. The impact of chronic migraine: the Chronic Migraine Epidemiology and Outcomes (CaMEO) Study methods and baseline results. Cephalalgia. 2015;35(7):563–78.PubMedPubMedCentralCrossRef
5.
go back to reference Arslantas D, Tozun M, Unsal A, Ozbek Z. Headache and its effects on health-related quality of life among adults. Turk Neurosurg. 2013;23(4):498–504.PubMed Arslantas D, Tozun M, Unsal A, Ozbek Z. Headache and its effects on health-related quality of life among adults. Turk Neurosurg. 2013;23(4):498–504.PubMed
6.
go back to reference Stovner L, Hagen K, Jensen R, Katsarava Z, Lipton R, et al. The global burden of headache: a documentation of headache prevalence and disability worldwide. Cephalalgia. 2007;27:193–210.PubMedCrossRef Stovner L, Hagen K, Jensen R, Katsarava Z, Lipton R, et al. The global burden of headache: a documentation of headache prevalence and disability worldwide. Cephalalgia. 2007;27:193–210.PubMedCrossRef
7.
go back to reference Russell MB, Ulrich V, Gervil M, Olesen J. Migraine without aura and migraine with aura are distinct disorders: a population-based twin survey. Headache. 2002;42:332–6.PubMedCrossRef Russell MB, Ulrich V, Gervil M, Olesen J. Migraine without aura and migraine with aura are distinct disorders: a population-based twin survey. Headache. 2002;42:332–6.PubMedCrossRef
8.
go back to reference Russell MB, Rasmussen BK, Fenger K, Olesen J. Migraine without aura and migraine with aura are distinct clinical entities: a study of four hundred and eighty-four male and female migraineurs from the general population. Cephalalgia. 1996;16:239–45.PubMedCrossRef Russell MB, Rasmussen BK, Fenger K, Olesen J. Migraine without aura and migraine with aura are distinct clinical entities: a study of four hundred and eighty-four male and female migraineurs from the general population. Cephalalgia. 1996;16:239–45.PubMedCrossRef
9.
go back to reference Headache Classification Subcommittee of the International Headache Society. The international classification of headache disorders, 3rd Edition (beta version). Cephalalgia. 2013;33(9):629–808.CrossRef Headache Classification Subcommittee of the International Headache Society. The international classification of headache disorders, 3rd Edition (beta version). Cephalalgia. 2013;33(9):629–808.CrossRef
10.
go back to reference Colson NJ, Lea RA, Quinlan S, Griffiths LR. The role of vascular and hormonal genes in migraine susceptibility. Mol Genet Metab. 2006;88(2):107–13.PubMedCrossRef Colson NJ, Lea RA, Quinlan S, Griffiths LR. The role of vascular and hormonal genes in migraine susceptibility. Mol Genet Metab. 2006;88(2):107–13.PubMedCrossRef
11.
go back to reference Fernandez F, Colson NJ, Griffiths LR. Pharmacogenetics of migraine: genetic variants and their potential role in migraine therapy. Pharmacogenomics. 2000;8(6):609 (14).CrossRef Fernandez F, Colson NJ, Griffiths LR. Pharmacogenetics of migraine: genetic variants and their potential role in migraine therapy. Pharmacogenomics. 2000;8(6):609 (14).CrossRef
12.
go back to reference Maher BH, Griffiths LR. Identification of molecular genetic factors that influence migraine. Mol Genet Genomics. 2011;285(6):433–46.PubMedCrossRef Maher BH, Griffiths LR. Identification of molecular genetic factors that influence migraine. Mol Genet Genomics. 2011;285(6):433–46.PubMedCrossRef
13.
go back to reference Colson N, Fernandez F, Griffiths L. Genetics of menstrual migraine: the molecular evidence. Curr Pain Headache Rep. 2010;14(5):389–95.PubMedCrossRef Colson N, Fernandez F, Griffiths L. Genetics of menstrual migraine: the molecular evidence. Curr Pain Headache Rep. 2010;14(5):389–95.PubMedCrossRef
14.
go back to reference Esserlind AL, Christensen AF, Le H, Kirchmann M, Hauge AW, et al. Replication and meta-analysis of common variants identifies a genome-wide significant locus in migraine. Eur J Neurol. 2013;20(5):765–72.PubMedCrossRef Esserlind AL, Christensen AF, Le H, Kirchmann M, Hauge AW, et al. Replication and meta-analysis of common variants identifies a genome-wide significant locus in migraine. Eur J Neurol. 2013;20(5):765–72.PubMedCrossRef
15.
go back to reference Chasman DI, Schürks M, Anttila V, Vries B, Schminke U, et al. Genome-wide association study reveals three susceptibility loci for common migraine in the general population. Nat Genet. 2011;43(7):695–8.PubMedPubMedCentralCrossRef Chasman DI, Schürks M, Anttila V, Vries B, Schminke U, et al. Genome-wide association study reveals three susceptibility loci for common migraine in the general population. Nat Genet. 2011;43(7):695–8.PubMedPubMedCentralCrossRef
16.
go back to reference Anttila V, Stefansson H, Kallela M, Todt U, Terwindt GM, et al. Genome-wide association study of migraine implicates a common susceptibility variant on 8q22.1. Nat Genet. 2010;42(10):869–73.PubMedPubMedCentralCrossRef Anttila V, Stefansson H, Kallela M, Todt U, Terwindt GM, et al. Genome-wide association study of migraine implicates a common susceptibility variant on 8q22.1. Nat Genet. 2010;42(10):869–73.PubMedPubMedCentralCrossRef
17.
go back to reference Gormley P, Anttila V, Winsvold BS, Palta P, Esko T, Pers TH, et al. Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat Genet. 2016;48:856–66.PubMedPubMedCentralCrossRef Gormley P, Anttila V, Winsvold BS, Palta P, Esko T, Pers TH, et al. Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat Genet. 2016;48:856–66.PubMedPubMedCentralCrossRef
18.
go back to reference Anttila V, Winsvold BS, Gormley P, Kurth T, Bettella F, McMahon G, et al. Genome-wide meta-analysis identifies new susceptibility loci for migraine. Nat Genet. 2013;45:912–7.PubMedPubMedCentralCrossRef Anttila V, Winsvold BS, Gormley P, Kurth T, Bettella F, McMahon G, et al. Genome-wide meta-analysis identifies new susceptibility loci for migraine. Nat Genet. 2013;45:912–7.PubMedPubMedCentralCrossRef
19.
go back to reference Pickrell JK, Berisa T, Liu JZ, Ségurel L, Tung JY, Hinds DA. Detection and interpretation of shared genetic influences on 42 human traits. Nat Genet. 2016;48:709–17.PubMedPubMedCentralCrossRef Pickrell JK, Berisa T, Liu JZ, Ségurel L, Tung JY, Hinds DA. Detection and interpretation of shared genetic influences on 42 human traits. Nat Genet. 2016;48:709–17.PubMedPubMedCentralCrossRef
20.
go back to reference Klimov E, Kondratieva N, Anuchina A, Skorobogatykh K, Azimova J, et al. Genetics of migraine - is there any progress? J Neurol Stroke. 2017;7(4):00245.CrossRef Klimov E, Kondratieva N, Anuchina A, Skorobogatykh K, Azimova J, et al. Genetics of migraine - is there any progress? J Neurol Stroke. 2017;7(4):00245.CrossRef
21.
go back to reference Goadsby PJ, Classey JD. Glutamatergic transmission in the trigeminal nucleus assessed with local blood flow. Brain Res. 2000;875:119–24.PubMedCrossRef Goadsby PJ, Classey JD. Glutamatergic transmission in the trigeminal nucleus assessed with local blood flow. Brain Res. 2000;875:119–24.PubMedCrossRef
22.
go back to reference Van den Maagdenberg AM, Haan J, Terwindt GM, Ferrari MD. Migraine: gene mutations and functional consequences. CurrOpinNeurol. 2007;20:299–305. Van den Maagdenberg AM, Haan J, Terwindt GM, Ferrari MD. Migraine: gene mutations and functional consequences. CurrOpinNeurol. 2007;20:299–305.
23.
go back to reference Martinez F, Castillo J, Rodriguez JR, Leira R, Noya M, et al. Neuroexcitatory amino acid levels in plasma and cerebrospinal fluid during migraine attacks. Cephalalgia. 1993;13:89–93.PubMedCrossRef Martinez F, Castillo J, Rodriguez JR, Leira R, Noya M, et al. Neuroexcitatory amino acid levels in plasma and cerebrospinal fluid during migraine attacks. Cephalalgia. 1993;13:89–93.PubMedCrossRef
25.
go back to reference Lauritzen M. Pathophysiology of the migraine aura: the spreading depression theory. Brain. 1994;117(Pt 1):199–210.PubMedCrossRef Lauritzen M. Pathophysiology of the migraine aura: the spreading depression theory. Brain. 1994;117(Pt 1):199–210.PubMedCrossRef
26.
go back to reference Andreou AP, Goadsby PJ. Therapeutic potential of novel glutamate receptor antagonists in migraine. Expert Opin Invest Drugs. 2009;18(6):789–803.CrossRef Andreou AP, Goadsby PJ. Therapeutic potential of novel glutamate receptor antagonists in migraine. Expert Opin Invest Drugs. 2009;18(6):789–803.CrossRef
27.
go back to reference Greenamyre JT, Young AB, Penney J. Quantitative autoradiographic distribution of l-[3H] glutamate-binding sites in rat central nervous system. J Neurosci. 1984;4(8):2133–44.PubMedCrossRef Greenamyre JT, Young AB, Penney J. Quantitative autoradiographic distribution of l-[3H] glutamate-binding sites in rat central nervous system. J Neurosci. 1984;4(8):2133–44.PubMedCrossRef
28.
go back to reference Kai–Kai MA, Howe R. Glutamate-immunoreactivity in the trigeminal and dorsal root ganglia, and intraspinal neurons and fibers in the dorsal horn of the rat. Histochem J. 1991;23(4):171–9.PubMedCrossRef Kai–Kai MA, Howe R. Glutamate-immunoreactivity in the trigeminal and dorsal root ganglia, and intraspinal neurons and fibers in the dorsal horn of the rat. Histochem J. 1991;23(4):171–9.PubMedCrossRef
29.
go back to reference Peres MF, Zukerman E, SenneSoares CA, Alonso EO, Santos BF, et al. Cerebrospinal fluid glutamate levels in chronic migraine. Cephalalgia. 2004;24(9):735–9.PubMedCrossRef Peres MF, Zukerman E, SenneSoares CA, Alonso EO, Santos BF, et al. Cerebrospinal fluid glutamate levels in chronic migraine. Cephalalgia. 2004;24(9):735–9.PubMedCrossRef
30.
go back to reference Rothrock JF, Mar KR, Yaksh TL, Golbeck A, Moore AC. Cerebrospinal fluid analyses in migraine patients and controls. Cephalalgia. 1995;15(6):489–93.PubMedCrossRef Rothrock JF, Mar KR, Yaksh TL, Golbeck A, Moore AC. Cerebrospinal fluid analyses in migraine patients and controls. Cephalalgia. 1995;15(6):489–93.PubMedCrossRef
31.
go back to reference Goyette P, Summer JS, Milos R, Ducan AM, Rosenblatt DS, et al. Human methylenetetrahydrofolatereductase isolation of cDNA, mapping and mutation identification. Nat Genet. 1994;7:195–200.PubMedCrossRef Goyette P, Summer JS, Milos R, Ducan AM, Rosenblatt DS, et al. Human methylenetetrahydrofolatereductase isolation of cDNA, mapping and mutation identification. Nat Genet. 1994;7:195–200.PubMedCrossRef
32.
go back to reference Frosst P, Blom HJ, Milos R, Goyette P, Sheppard CA, et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydofolatereductase. Nat Genet. 1995;10:111–3.PubMedCrossRef Frosst P, Blom HJ, Milos R, Goyette P, Sheppard CA, et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydofolatereductase. Nat Genet. 1995;10:111–3.PubMedCrossRef
34.
go back to reference Coull BM, Malinow MR, Beamer N, Sexton G, Nordt F, et al. Elevated plasma homocysteine concentrations as a possible independent risk factor for stroke. Stroke. 1990;21:572–6.PubMedCrossRef Coull BM, Malinow MR, Beamer N, Sexton G, Nordt F, et al. Elevated plasma homocysteine concentrations as a possible independent risk factor for stroke. Stroke. 1990;21:572–6.PubMedCrossRef
35.
go back to reference Kelly PJ, Rosand J, Kistler JP, Shih VE, Silveira S, et al. Homocysteine, MTHFR 677C-->T polymorphism, and risk of ischemic stroke: results of a meta-analysis. Neurology. 2002;59:529–36.PubMedCrossRef Kelly PJ, Rosand J, Kistler JP, Shih VE, Silveira S, et al. Homocysteine, MTHFR 677C-->T polymorphism, and risk of ischemic stroke: results of a meta-analysis. Neurology. 2002;59:529–36.PubMedCrossRef
36.
go back to reference Schwaag S, Nabavi DG, Frese A, Husstedt IW, Evers S. The association between migraine and juvenile stroke: a case-control study. Headache. 2003;43:90–5.PubMedCrossRef Schwaag S, Nabavi DG, Frese A, Husstedt IW, Evers S. The association between migraine and juvenile stroke: a case-control study. Headache. 2003;43:90–5.PubMedCrossRef
37.
go back to reference Donaghy M, Chang CL, Poulter NJ. Duration, frequency, recency, and type of migraine and the risk of ischaemic stroke in women of childbearing age. NeurolNeurosurg Psychiatry. 2002;73:747–50.CrossRef Donaghy M, Chang CL, Poulter NJ. Duration, frequency, recency, and type of migraine and the risk of ischaemic stroke in women of childbearing age. NeurolNeurosurg Psychiatry. 2002;73:747–50.CrossRef
38.
go back to reference Tzourio C, Iglesias S, Hubert JB, Visy JM, Alperovitch A, et al. Migraine and risk of ischaemic stroke: a case-control study. PMID. 1993;31:289–92. Tzourio C, Iglesias S, Hubert JB, Visy JM, Alperovitch A, et al. Migraine and risk of ischaemic stroke: a case-control study. PMID. 1993;31:289–92.
39.
go back to reference Weisberg IS, Jacques FP, Selhub J, Bostom AG, Chen Z, et al. The 1298A→C polymorphism in methylenetetrahydrofolatereductase (MTHFR): in vitro expression and association with homocysteine. Atherosclerosis. 2001;156:409–15.PubMedCrossRef Weisberg IS, Jacques FP, Selhub J, Bostom AG, Chen Z, et al. The 1298A→C polymorphism in methylenetetrahydrofolatereductase (MTHFR): in vitro expression and association with homocysteine. Atherosclerosis. 2001;156:409–15.PubMedCrossRef
40.
go back to reference Friedman G, Goldschmidt N, Friedlander Y, Ben-Yehuda A, Selhub J, et al. A common mutation A1298C in human methylenetetrahydrofolatereductase gene: association with plasma total homocysteine and folate concentrations. JNutr. 1999;129(9):1656–61. Friedman G, Goldschmidt N, Friedlander Y, Ben-Yehuda A, Selhub J, et al. A common mutation A1298C in human methylenetetrahydrofolatereductase gene: association with plasma total homocysteine and folate concentrations. JNutr. 1999;129(9):1656–61.
41.
go back to reference Lillis AP, Duyn LB, Murphy-Ullrich JE, Strickland DK. LDL receptor-related protein 1: unique tissue-specific functions revealed by selective gene knockout studies. Physiol Rev. 2008;88(3):887–918.PubMedPubMedCentralCrossRef Lillis AP, Duyn LB, Murphy-Ullrich JE, Strickland DK. LDL receptor-related protein 1: unique tissue-specific functions revealed by selective gene knockout studies. Physiol Rev. 2008;88(3):887–918.PubMedPubMedCentralCrossRef
42.
go back to reference Proudfoot CJ, Garry EM, Cottrell DF, Rosie R, Anderson H, et al. Analgesia mediated by the TRPM8 cold receptor in chronic neuropathic pain. CurrBiol. 2006;16(16):1591–605.CrossRef Proudfoot CJ, Garry EM, Cottrell DF, Rosie R, Anderson H, et al. Analgesia mediated by the TRPM8 cold receptor in chronic neuropathic pain. CurrBiol. 2006;16(16):1591–605.CrossRef
43.
go back to reference Peier AM, Moqrich A, Hergarden AC, Reeve AJ, Andersson DA, et al. A TRP channel that senses cold stimuli and menthol. Cell. 2002;108(5):705–15.PubMedCrossRef Peier AM, Moqrich A, Hergarden AC, Reeve AJ, Andersson DA, et al. A TRP channel that senses cold stimuli and menthol. Cell. 2002;108(5):705–15.PubMedCrossRef
45.
46.
go back to reference Seale P, Bjork B, Yang W, Kajimura S, Chin S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;15:961–7.CrossRef Seale P, Bjork B, Yang W, Kajimura S, Chin S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;15:961–7.CrossRef
47.
go back to reference Takahata M, Inoue Y, Tsuda H, Imoto I, Koinuma D, et al. SKI and MEL1 cooperate to inhibit transforming growth factor-beta signal in gastric cancer cells. J BiolChem. 2009;15:3334–44. Takahata M, Inoue Y, Tsuda H, Imoto I, Koinuma D, et al. SKI and MEL1 cooperate to inhibit transforming growth factor-beta signal in gastric cancer cells. J BiolChem. 2009;15:3334–44.
48.
go back to reference Sherman EA, Strauss KA, Tortorelli S, Bennett MJ, Knerr I, et al. Genetic mapping of glutaricaciduria, type 3, to chromosome 7 and identification of mutations in c7orf10. Am J Hum Genet. 2008;83:604–9.PubMedPubMedCentralCrossRef Sherman EA, Strauss KA, Tortorelli S, Bennett MJ, Knerr I, et al. Genetic mapping of glutaricaciduria, type 3, to chromosome 7 and identification of mutations in c7orf10. Am J Hum Genet. 2008;83:604–9.PubMedPubMedCentralCrossRef
49.
go back to reference Marlaire S, Van Schaftingen E, Veiga-da-Cunha M. C7orf10 encodes succinate-hydroxymethylglutarate CoA-transferase, the enzyme that converts glutarate to glutaryl-CoA. J Inherit Metab Dis. 2004;37(1):13–9.CrossRef Marlaire S, Van Schaftingen E, Veiga-da-Cunha M. C7orf10 encodes succinate-hydroxymethylglutarate CoA-transferase, the enzyme that converts glutarate to glutaryl-CoA. J Inherit Metab Dis. 2004;37(1):13–9.CrossRef
50.
go back to reference Rozanov DV, Hahn-Dantona E, Strickland DK, Strongin AY. The low density lipoprotein receptor-related protein LRP is regulated by membrane type-1 matrix metalloproteinase (MT1-MMP) proteolysis in malignant cells. J BiolChem. 2004;279:4260–8. Rozanov DV, Hahn-Dantona E, Strickland DK, Strongin AY. The low density lipoprotein receptor-related protein LRP is regulated by membrane type-1 matrix metalloproteinase (MT1-MMP) proteolysis in malignant cells. J BiolChem. 2004;279:4260–8.
51.
go back to reference Borrie SC, Baeumer BE, Bandtlow CE. The Nogo-66 receptor family in the intact and diseased CNS. Cell Tissue Res. 2012;349:105–17.PubMedCrossRef Borrie SC, Baeumer BE, Bandtlow CE. The Nogo-66 receptor family in the intact and diseased CNS. Cell Tissue Res. 2012;349:105–17.PubMedCrossRef
52.
go back to reference Schreiner A, Ruonala M, Jakob V, Suthaus J, Boles E, et al. Junction protein shrew-1 influences cell invasion and interacts with invasion-promoting protein CD147. MolBiol Cell. 2007;18:1272–81. Schreiner A, Ruonala M, Jakob V, Suthaus J, Boles E, et al. Junction protein shrew-1 influences cell invasion and interacts with invasion-promoting protein CD147. MolBiol Cell. 2007;18:1272–81.
53.
go back to reference Lafleur MA, Xu D, Hemler ME. Tetraspanin proteins regulate membrane type-1 matrix metalloproteinase-dependent pericellular proteolysis. MolBiol Cell. 2009;20(7):2030–40. Lafleur MA, Xu D, Hemler ME. Tetraspanin proteins regulate membrane type-1 matrix metalloproteinase-dependent pericellular proteolysis. MolBiol Cell. 2009;20(7):2030–40.
54.
go back to reference Fimia GM, De Cesare D, Sassone-Corsi P. A family of LIM-only transcriptional coactivators: tissue-specific expression and selective activation of CREB and CREM. Mol Cell Biol. 2000;20:8613–22.PubMedPubMedCentralCrossRef Fimia GM, De Cesare D, Sassone-Corsi P. A family of LIM-only transcriptional coactivators: tissue-specific expression and selective activation of CREB and CREM. Mol Cell Biol. 2000;20:8613–22.PubMedPubMedCentralCrossRef
55.
go back to reference Dash PK, Hochner B, Kandel ER. Injection of the cAMP-responsive element into the nucleus of Aplysia sensory neurons blocks long-term facilitation. Nature. 1990;345:718–21.PubMedCrossRef Dash PK, Hochner B, Kandel ER. Injection of the cAMP-responsive element into the nucleus of Aplysia sensory neurons blocks long-term facilitation. Nature. 1990;345:718–21.PubMedCrossRef
57.
go back to reference Freilinger T, Anttila V, De VB, Malik R, Kallela M, et al. Genome-wide association analysis identifies susceptibility loci for migraine without aura. Nat Genet. 2012;15:777–82.CrossRef Freilinger T, Anttila V, De VB, Malik R, Kallela M, et al. Genome-wide association analysis identifies susceptibility loci for migraine without aura. Nat Genet. 2012;15:777–82.CrossRef
58.
go back to reference Flavell SW, Cowan CW, Kim TK, Greer PL, Lin Y, et al. Activity-dependent regulation of MEF2 transcription factors suppresses excitatory synapse number. Science. 2006;15:1008–12.CrossRef Flavell SW, Cowan CW, Kim TK, Greer PL, Lin Y, et al. Activity-dependent regulation of MEF2 transcription factors suppresses excitatory synapse number. Science. 2006;15:1008–12.CrossRef
59.
go back to reference Allen PB, Greenfield AT, Svenningsson P, Haspeslagh DC, Greengard P. Phactrs 1–4: a family of protein phosphatase 1 and actin regulatory proteins. ProcNatlAcadSci U S A. 2004;15:7187–92.CrossRef Allen PB, Greenfield AT, Svenningsson P, Haspeslagh DC, Greengard P. Phactrs 1–4: a family of protein phosphatase 1 and actin regulatory proteins. ProcNatlAcadSci U S A. 2004;15:7187–92.CrossRef
60.
go back to reference Greengard P, Allen PB, Nairn AC. Beyond the dopamine receptor: the DARPP-32/protein phosphatase-1 cascade. Neuron. 1999;15:435–47.CrossRef Greengard P, Allen PB, Nairn AC. Beyond the dopamine receptor: the DARPP-32/protein phosphatase-1 cascade. Neuron. 1999;15:435–47.CrossRef
61.
go back to reference Lin HY, Wang XF, Ng-Eaton E, Weinberg RA, Lodish HF. Expression cloning of the TGF-beta type II receptor, a functional transmembrane serine/threonine kinase. Cell. 1992;15:775–85.CrossRef Lin HY, Wang XF, Ng-Eaton E, Weinberg RA, Lodish HF. Expression cloning of the TGF-beta type II receptor, a functional transmembrane serine/threonine kinase. Cell. 1992;15:775–85.CrossRef
62.
go back to reference Wilson PM, Fryer RH, Fang Y, Hatten ME. Astn2, a novel member of the astrotactin gene family, regulates the trafficking of ASTN1 during glial-guided neuronal migration. J Neurosci. 2010;15:8529–40.CrossRef Wilson PM, Fryer RH, Fang Y, Hatten ME. Astn2, a novel member of the astrotactin gene family, regulates the trafficking of ASTN1 during glial-guided neuronal migration. J Neurosci. 2010;15:8529–40.CrossRef
63.
go back to reference Schürks M, Rist PM, Bigal ME, Buring JE, Lipton RB, et al. Migraine and cardiovascular disease: systematic review and meta analysis. BMJ. 2009;339:b3914.PubMedPubMedCentralCrossRef Schürks M, Rist PM, Bigal ME, Buring JE, Lipton RB, et al. Migraine and cardiovascular disease: systematic review and meta analysis. BMJ. 2009;339:b3914.PubMedPubMedCentralCrossRef
64.
65.
go back to reference Biros E, Walker PJ, Nataatmadja M, West M, Golledge J. Downregulation of transforming growth factor, beta receptor 2 and Notch signaling pathway in human abdominal aorticaneurysm. Atherosclerosis. 2012;221:383–6.PubMedCrossRef Biros E, Walker PJ, Nataatmadja M, West M, Golledge J. Downregulation of transforming growth factor, beta receptor 2 and Notch signaling pathway in human abdominal aorticaneurysm. Atherosclerosis. 2012;221:383–6.PubMedCrossRef
66.
go back to reference Kathiresan S, Voight BF, Purcell S, Musunuru K, Ardissino D, et al. Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat Genet. 2009;41:334–41.PubMedCrossRef Kathiresan S, Voight BF, Purcell S, Musunuru K, Ardissino D, et al. Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat Genet. 2009;41:334–41.PubMedCrossRef
67.
go back to reference Terada N, Baracskay K, Kinter M, Melrose S, Brophy PJ, et al. The tetraspanin protein, CD9, is expressed by progenitor cells committed to oligodendrogenesis and is linked to beta1 integrin, CD81, and Tspan-2. Glia. 2002;40:350–9.PubMedCrossRef Terada N, Baracskay K, Kinter M, Melrose S, Brophy PJ, et al. The tetraspanin protein, CD9, is expressed by progenitor cells committed to oligodendrogenesis and is linked to beta1 integrin, CD81, and Tspan-2. Glia. 2002;40:350–9.PubMedCrossRef
68.
go back to reference Flavell SW, Kim TK, Gray JM, Harmin DA, Hemberg M, et al. Genome-wide analysis of MEF2 transcriptional program reveals synaptic target genes and neuronal activity-dependent polyadenylation site selection. Neuron. 2008;60:1022–38.PubMedPubMedCentralCrossRef Flavell SW, Kim TK, Gray JM, Harmin DA, Hemberg M, et al. Genome-wide analysis of MEF2 transcriptional program reveals synaptic target genes and neuronal activity-dependent polyadenylation site selection. Neuron. 2008;60:1022–38.PubMedPubMedCentralCrossRef
69.
go back to reference Tsuzuki K, Xing H, Ling J, Gu JG. Menthol-induced Ca2+ release from presynaptic Ca2+ stores potentiates sensory synaptic transmission. J Neurosci. 2004;24:762–71.PubMedCrossRef Tsuzuki K, Xing H, Ling J, Gu JG. Menthol-induced Ca2+ release from presynaptic Ca2+ stores potentiates sensory synaptic transmission. J Neurosci. 2004;24:762–71.PubMedCrossRef
70.
go back to reference Diniz LP, Almeida JC, Tortelli V, Vargas Lopes C, Setti-Perdigão P, et al. Astrocyte-induced synaptogenesis is mediated by transforming growth factor beta signaling through modulation of D-serine levels in cerebral cortex neuron. J BiolChem. 2012;287(49):41432–41,445. Diniz LP, Almeida JC, Tortelli V, Vargas Lopes C, Setti-Perdigão P, et al. Astrocyte-induced synaptogenesis is mediated by transforming growth factor beta signaling through modulation of D-serine levels in cerebral cortex neuron. J BiolChem. 2012;287(49):41432–41,445.
71.
go back to reference Allen PB, Greenfield AT, Svenningsson P, Haspeslagh DC, Greengard P. Phactrs. 1–4: a family of protein phosphatase 1 and actin regulatory proteins. ProcNatlAcad Sci USA. 2004;101:7187–92.CrossRef Allen PB, Greenfield AT, Svenningsson P, Haspeslagh DC, Greengard P. Phactrs. 1–4: a family of protein phosphatase 1 and actin regulatory proteins. ProcNatlAcad Sci USA. 2004;101:7187–92.CrossRef
72.
go back to reference Ferraro GB, Morrison CJ, Overall CM, Strittmatter SM, Fournier AE. Membrane-type matrix metalloproteinase-3 regulates neuronal responsiveness to myelin through Nogo-66 receptor 1 cleavage. J BiolChem. 2011;286:31418–24. Ferraro GB, Morrison CJ, Overall CM, Strittmatter SM, Fournier AE. Membrane-type matrix metalloproteinase-3 regulates neuronal responsiveness to myelin through Nogo-66 receptor 1 cleavage. J BiolChem. 2011;286:31418–24.
73.
go back to reference May P, Rohlmann A, Bock HH, Zurhove K, Marth JD, et al. Neuronal LRP1 functionally associates with postsynaptic proteins and is required for normal motor function in mice. Mol Cell Biol. 2004;24:8872–83.PubMedPubMedCentralCrossRef May P, Rohlmann A, Bock HH, Zurhove K, Marth JD, et al. Neuronal LRP1 functionally associates with postsynaptic proteins and is required for normal motor function in mice. Mol Cell Biol. 2004;24:8872–83.PubMedPubMedCentralCrossRef
Metadata
Title
Role of single nucleotide polymorphisms (SNPs) in common migraine
Authors
Sukhvinder Kaur
Arif Ali
Uzair Ahmad
Yaser Siahbalaei
A. K. Pandey
Balkirat Singh
Publication date
01-12-2019
Publisher
Springer Berlin Heidelberg
Keywords
Migraine
Aura
DOI
https://doi.org/10.1186/s41983-019-0093-8

Other articles of this Issue 1/2019

The Egyptian Journal of Neurology, Psychiatry and Neurosurgery 1/2019 Go to the issue