Skip to main content
Top
Published in: The Egyptian Journal of Neurology, Psychiatry and Neurosurgery 1/2021

Open Access 01-12-2021 | Migraine | Research

A volumetric magnetic resonance imaging study in migraine

Authors: Laila Elmously Naguib, Ghada Saed Abdel Azim, Mohammed Abdelrazek Abdellatif

Published in: The Egyptian Journal of Neurology, Psychiatry and Neurosurgery | Issue 1/2021

Login to get access

Abstract

Background

Although migraine phenotype has been widely described, the explanation of migraine pathophysiology still has a gap that might be partly bridged by neuroimaging investigations. The aim of the study is to assess volumetric brain changes in migraineurs compared with controls, and in episodic migraine in comparison to chronic type. Structural brain changes in migraineurs (with and without aura) were assessed by an automated segmentation method (Free Surfer). T1-weighted MRIs of 25 migraineurs (14 diagnosed as episodic type and 11 diagnosed as chronic migraine) and 25 headache-free controls were evaluated and processed.

Results

Migraine patients had significant reduction of the volume of total brain, grey matter, brain stem, cerebellum, basal ganglia, thalamus, hippocampus and amygdala in comparison to control subjects. Patients with chronic migraine had significant reduction in volume of total brain, grey matter, cerebellum and frontal lobe thickness in comparison to those with episodic migraine.

Conclusion

Migraineurs showed volumetric brain changes mainly in areas related to central processing of pain and in areas specific for migraine (such as brain stem) when compared to healthy controls. Chronic migraineurs showed significant reduction in grey matter, in areas involved in processing of pain, cognition and multisensory integration versus patients with episodic migraine, which adds insight into the pathophysiology of migraine as a progressive disorder that may have long-term impacts on the brain as regards structure and function.
Literature
1.
go back to reference Bashir A, Lipton RB, Ashina S, Ashina M. Migraine and structural changes in the brain: a systematic review and meta-analysis. Neurology. 2013;81(14):1260–8. PubMedPubMedCentralCrossRef Bashir A, Lipton RB, Ashina S, Ashina M. Migraine and structural changes in the brain: a systematic review and meta-analysis. Neurology. 2013;81(14):1260–8. PubMedPubMedCentralCrossRef
2.
3.
go back to reference Jin C, Yuan K, Zhao L, Zhao L, Yu D, von Deneen KM, et al. Structural and functional abnormalities in migraine patients without aura. NMR Biomed. 2013;26(1):58–64.PubMedCrossRef Jin C, Yuan K, Zhao L, Zhao L, Yu D, von Deneen KM, et al. Structural and functional abnormalities in migraine patients without aura. NMR Biomed. 2013;26(1):58–64.PubMedCrossRef
4.
go back to reference Bigal ME, Lipton RB. What predicts the change from episodic to chronic migraine? Curr Opin Neurol. 2009;22:269–76.PubMedCrossRef Bigal ME, Lipton RB. What predicts the change from episodic to chronic migraine? Curr Opin Neurol. 2009;22:269–76.PubMedCrossRef
5.
go back to reference Mathew NT. Pathophysiology of chronic migraine and mode of action of preventive medications. Headache. 2011;51(Suppl 2):84–92.PubMedCrossRef Mathew NT. Pathophysiology of chronic migraine and mode of action of preventive medications. Headache. 2011;51(Suppl 2):84–92.PubMedCrossRef
6.
go back to reference Headache Classification Committee of the International Headache Society (IHS). The international classification of headache disorders (beta version). Cephalalgia. 2013;33(9):629–808.CrossRef Headache Classification Committee of the International Headache Society (IHS). The international classification of headache disorders (beta version). Cephalalgia. 2013;33(9):629–808.CrossRef
7.
go back to reference Bagley L, Rendas-Baum R, Gregory A, Yang M, Varon S, Lee J, et al. Validating migraine specific quality of life questionnaire. Headache. 2012;52(3):409–21.CrossRefPubMed Bagley L, Rendas-Baum R, Gregory A, Yang M, Varon S, Lee J, et al. Validating migraine specific quality of life questionnaire. Headache. 2012;52(3):409–21.CrossRefPubMed
8.
go back to reference Beck T, Steer A, Brown K. Manual for the beck depression inventory–II. San Antonio: Psychological Corporation; 1996. Beck T, Steer A, Brown K. Manual for the beck depression inventory–II. San Antonio: Psychological Corporation; 1996.
9.
go back to reference Mansbach W, Mac Dougall E, Rosenzweig S. The Brief Cognitive Assessment Tool (BCAT): a new test emphasizing contextual memory, executive functions, attentional capacity, and the prediction of instrumental activities of daily living. J Clin Exp Neuropsychol. 2012;34(2):183–94.PubMedCrossRef Mansbach W, Mac Dougall E, Rosenzweig S. The Brief Cognitive Assessment Tool (BCAT): a new test emphasizing contextual memory, executive functions, attentional capacity, and the prediction of instrumental activities of daily living. J Clin Exp Neuropsychol. 2012;34(2):183–94.PubMedCrossRef
10.
go back to reference Reuter M, Rosas D, Fischl B. Highly accurate inverse consistent registration: a robust approach. Neuroimage. 2010;53(4):1181–96.PubMedCrossRef Reuter M, Rosas D, Fischl B. Highly accurate inverse consistent registration: a robust approach. Neuroimage. 2010;53(4):1181–96.PubMedCrossRef
11.
go back to reference Segonne F, Dale M, Busa E, Glessner M, Salat D, Hahn K, et al. A hybrid approach to the skull stripping problem in MRI. Neuroimage. 2004;22:1060–75.PubMedCrossRef Segonne F, Dale M, Busa E, Glessner M, Salat D, Hahn K, et al. A hybrid approach to the skull stripping problem in MRI. Neuroimage. 2004;22:1060–75.PubMedCrossRef
12.
go back to reference Fischl B, Salat D, Busa E, Albert M, Dieterich M, Haselgrove C, et al. Whole brain segmentation: automated labeling of neuroanatomical structures in the human brain. Neuron. 2002;33:341–55.PubMedCrossRef Fischl B, Salat D, Busa E, Albert M, Dieterich M, Haselgrove C, et al. Whole brain segmentation: automated labeling of neuroanatomical structures in the human brain. Neuron. 2002;33:341–55.PubMedCrossRef
13.
go back to reference Fischl B, Salat D, van der Kouwe A, Makris N, Ségonne F, Quinn T, et al. Sequence-independent segmentation of magnetic resonance images. Neuroimage. 2004;23(1):S69-84.PubMedCrossRef Fischl B, Salat D, van der Kouwe A, Makris N, Ségonne F, Quinn T, et al. Sequence-independent segmentation of magnetic resonance images. Neuroimage. 2004;23(1):S69-84.PubMedCrossRef
14.
go back to reference Sled J, Zijdenbos P, Evans C. A nonparametric method for automatic correction of intensity nonuniformity in MRI data. IEEE Trans Med Imaging. 1998;17:87–97.PubMedCrossRef Sled J, Zijdenbos P, Evans C. A nonparametric method for automatic correction of intensity nonuniformity in MRI data. IEEE Trans Med Imaging. 1998;17:87–97.PubMedCrossRef
15.
go back to reference Fischl B, Liu A, Dale M. Automated manifold surgery: constructing geometrically accurate and topologically correct models of the human cerebral cortex. IEEE Trans Med Imaging. 2001;20:70–80.PubMedCrossRef Fischl B, Liu A, Dale M. Automated manifold surgery: constructing geometrically accurate and topologically correct models of the human cerebral cortex. IEEE Trans Med Imaging. 2001;20:70–80.PubMedCrossRef
16.
go back to reference Segonne F, Pacheco J, Fischl B. Geometrically accurate topology-correction of cortical surfaces using nonseparating loops. IEEE Trans Med Imaging. 2007;26:518529.CrossRef Segonne F, Pacheco J, Fischl B. Geometrically accurate topology-correction of cortical surfaces using nonseparating loops. IEEE Trans Med Imaging. 2007;26:518529.CrossRef
17.
go back to reference Dale M, Fischl B, Sereno I. Cortical surface-based analysis. I. Segmentation and surface reconstruction. Neuroimage. 1999;9:179–94.PubMedCrossRef Dale M, Fischl B, Sereno I. Cortical surface-based analysis. I. Segmentation and surface reconstruction. Neuroimage. 1999;9:179–94.PubMedCrossRef
18.
go back to reference Dale M, Sereno I. Improved localization of cortical activity by combining EEG and MEG with MRI cortical surface reconstruction: a linear approach. J Cogn Neurosci. 1993;5:162–76.PubMedCrossRef Dale M, Sereno I. Improved localization of cortical activity by combining EEG and MEG with MRI cortical surface reconstruction: a linear approach. J Cogn Neurosci. 1993;5:162–76.PubMedCrossRef
19.
20.
go back to reference Gudmundsson S, Scher I, Sigurdsson S, Mirjam G, JeanSebastien V, Gudny E. Migraine, depression, and brain volume. The AGES-Reykjavik Study. Neurology. 2013;80(23):2138–44.PubMedPubMedCentralCrossRef Gudmundsson S, Scher I, Sigurdsson S, Mirjam G, JeanSebastien V, Gudny E. Migraine, depression, and brain volume. The AGES-Reykjavik Study. Neurology. 2013;80(23):2138–44.PubMedPubMedCentralCrossRef
22.
go back to reference Timmann D, Drepper J, Frings M, Maschke M, Richter S, Gerwig M, et al. The human cerebellum contributes to motor, emotional and cognitive associative learning. A review. Cortex. 2010;46(7):845–57.PubMedCrossRef Timmann D, Drepper J, Frings M, Maschke M, Richter S, Gerwig M, et al. The human cerebellum contributes to motor, emotional and cognitive associative learning. A review. Cortex. 2010;46(7):845–57.PubMedCrossRef
23.
go back to reference Moulton EA, Schmahmann JD, Becerra L, Borsook D. The cerebellum and pain: passive integrator or active participator? Brain Res Rev. 2010;65(1):14–27.PubMedPubMedCentralCrossRef Moulton EA, Schmahmann JD, Becerra L, Borsook D. The cerebellum and pain: passive integrator or active participator? Brain Res Rev. 2010;65(1):14–27.PubMedPubMedCentralCrossRef
24.
go back to reference Bocci T, Santarcangelo E, Vannini B, Torzini A, Carli G, Ferrucci R, et al. Cerebellar direct current stimulation modulates pain perception in humans. Restor Neurol Neurosci. 2015;33(5):597–609.PubMed Bocci T, Santarcangelo E, Vannini B, Torzini A, Carli G, Ferrucci R, et al. Cerebellar direct current stimulation modulates pain perception in humans. Restor Neurol Neurosci. 2015;33(5):597–609.PubMed
25.
go back to reference Pereira M, Rafiq B, Chowdhury E, Babayev J, Boo H, Metwaly R, et al. Anodal cerebellar tDCS modulates lower extremity pain perception. NeuroRehabilitation. 2017;40(2):195–200.PubMedCrossRef Pereira M, Rafiq B, Chowdhury E, Babayev J, Boo H, Metwaly R, et al. Anodal cerebellar tDCS modulates lower extremity pain perception. NeuroRehabilitation. 2017;40(2):195–200.PubMedCrossRef
26.
go back to reference Bocci T, De Carolis G, Ferrucci R, Paroli M, Mansani F, Priori A, et al. Cerebellar Transcranial direct current stimulation (ctDCS) ameliorates phantom limb pain and non-painful phantom limb sensations. Cerebellum. 2019;18(3):527–35.PubMedCrossRef Bocci T, De Carolis G, Ferrucci R, Paroli M, Mansani F, Priori A, et al. Cerebellar Transcranial direct current stimulation (ctDCS) ameliorates phantom limb pain and non-painful phantom limb sensations. Cerebellum. 2019;18(3):527–35.PubMedCrossRef
27.
go back to reference Kros L, AngueyraAristizábal CA, Khodakhah K. Cerebellar involvement in migraine. Cephalalgia. 2018;38(11):1782–91.PubMedCrossRef Kros L, AngueyraAristizábal CA, Khodakhah K. Cerebellar involvement in migraine. Cephalalgia. 2018;38(11):1782–91.PubMedCrossRef
28.
go back to reference Mehnert J, Schulte L, Timmann D, May A. Activity and connectivity of the cerebellum in trigeminal nociception. Neuroimage. 2017;150:112–8.PubMedCrossRef Mehnert J, Schulte L, Timmann D, May A. Activity and connectivity of the cerebellum in trigeminal nociception. Neuroimage. 2017;150:112–8.PubMedCrossRef
29.
go back to reference Koppen H, Boele H-J, Palm-Meinders IH, Koutstaal BJ, Horlings CGC, Koekkoek BK, et al. Cerebellar function and ischemic brain lesions in migraine patients from the general population. Cephalalgia. 2016;37(2):177–90.PubMedPubMedCentralCrossRef Koppen H, Boele H-J, Palm-Meinders IH, Koutstaal BJ, Horlings CGC, Koekkoek BK, et al. Cerebellar function and ischemic brain lesions in migraine patients from the general population. Cephalalgia. 2016;37(2):177–90.PubMedPubMedCentralCrossRef
30.
go back to reference Kruit MC, van Buchem MA, Hofman PA, Bakkers JT, Terwindt GM, Ferrari MD, et al. Migraine as a risk factor for subclinical brain lesions. JAMA. 2004;291(4):427–34.PubMedCrossRef Kruit MC, van Buchem MA, Hofman PA, Bakkers JT, Terwindt GM, Ferrari MD, et al. Migraine as a risk factor for subclinical brain lesions. JAMA. 2004;291(4):427–34.PubMedCrossRef
31.
go back to reference Liu HY, Lee PL, Chou KH, Lai KL, Wang YF, Chen SP, et al. The cerebellum is associated with 2-year prognosis in patients with high-frequency migraine. J Headache Pain. 2020;21(1):29.PubMedPubMedCentralCrossRef Liu HY, Lee PL, Chou KH, Lai KL, Wang YF, Chen SP, et al. The cerebellum is associated with 2-year prognosis in patients with high-frequency migraine. J Headache Pain. 2020;21(1):29.PubMedPubMedCentralCrossRef
33.
go back to reference Bilgiç B, Kocaman G, Arslan B, Noyan H, Sherifov R, Alkan A, et al. Volumetric differences suggest involvement of cerebellum and brainstem in chronic migraine. Cephalalgia. 2016;36(4):301–8.PubMedCrossRef Bilgiç B, Kocaman G, Arslan B, Noyan H, Sherifov R, Alkan A, et al. Volumetric differences suggest involvement of cerebellum and brainstem in chronic migraine. Cephalalgia. 2016;36(4):301–8.PubMedCrossRef
35.
go back to reference Kruit MC, Launer LJ, Overbosch J, van Buchem MA, Ferrari MD. Iron accumulation in deep brain nuclei in migraine: a population-based magnetic resonance imaging study. Cephalalgia. 2009;29:351–9.PubMedCrossRef Kruit MC, Launer LJ, Overbosch J, van Buchem MA, Ferrari MD. Iron accumulation in deep brain nuclei in migraine: a population-based magnetic resonance imaging study. Cephalalgia. 2009;29:351–9.PubMedCrossRef
36.
go back to reference Yuan K, Zhao L, Cheng P, Yu D, Zhao L, Dong T, et al. Altered structure and resting-state functional connectivity of the basal ganglia in migraine patients without aura. J Pain. 2013;14:836–44.PubMedCrossRef Yuan K, Zhao L, Cheng P, Yu D, Zhao L, Dong T, et al. Altered structure and resting-state functional connectivity of the basal ganglia in migraine patients without aura. J Pain. 2013;14:836–44.PubMedCrossRef
37.
go back to reference Rocca MA, Messina R, Colombo B, Falini A, Comi G, Filippi M. Structural brain MRI abnormalities in pediatric patients with migraine. J Neurol. 2014;261:350–7.PubMedCrossRef Rocca MA, Messina R, Colombo B, Falini A, Comi G, Filippi M. Structural brain MRI abnormalities in pediatric patients with migraine. J Neurol. 2014;261:350–7.PubMedCrossRef
38.
go back to reference Magon S, May A, Stankewitz A, Goadsby PJ, Tso AR, Ashina M, et al. Morphological abnormalities of thalamic subnuclei in migraine: a multicenter MRI study at 3 Tesla. J Neurosci. 2015;35:13800–6.PubMedPubMedCentralCrossRef Magon S, May A, Stankewitz A, Goadsby PJ, Tso AR, Ashina M, et al. Morphological abnormalities of thalamic subnuclei in migraine: a multicenter MRI study at 3 Tesla. J Neurosci. 2015;35:13800–6.PubMedPubMedCentralCrossRef
39.
go back to reference Schmitz N, Arkink EB, Mulder M, Rubia K, Admiraal-Behloul F, Schoonman GG, et al. Frontal lobe structure and executive function in migraine patients. Neurosci Lett. 2008;440:92–6.PubMedCrossRef Schmitz N, Arkink EB, Mulder M, Rubia K, Admiraal-Behloul F, Schoonman GG, et al. Frontal lobe structure and executive function in migraine patients. Neurosci Lett. 2008;440:92–6.PubMedCrossRef
40.
go back to reference Younis S, Hougaard A, Noseda R, Ashina M. Current understanding of thalamic structure and function in migraine. Cephalalgia. 2019;39:1675–82.PubMedCrossRef Younis S, Hougaard A, Noseda R, Ashina M. Current understanding of thalamic structure and function in migraine. Cephalalgia. 2019;39:1675–82.PubMedCrossRef
41.
go back to reference Granziera C, Daducci A, Romascano D, Roche A, Helms G, Krueger G, et al. Structural abnormalities in the thalamus of migraineurs with aura: a multiparametric study at 3 T. Hum Brain Mapp. 2014;35:1461–8.PubMedCrossRef Granziera C, Daducci A, Romascano D, Roche A, Helms G, Krueger G, et al. Structural abnormalities in the thalamus of migraineurs with aura: a multiparametric study at 3 T. Hum Brain Mapp. 2014;35:1461–8.PubMedCrossRef
42.
go back to reference Marciszewski KK, Meylakh N, Di Pietro F, Macefield VG, Macey PM, Henderson LA. Altered brainstem anatomy in migraine. Cephalalgia. 2018;38:476–86.PubMedCrossRef Marciszewski KK, Meylakh N, Di Pietro F, Macefield VG, Macey PM, Henderson LA. Altered brainstem anatomy in migraine. Cephalalgia. 2018;38:476–86.PubMedCrossRef
43.
44.
go back to reference Chen ZY, Chen XY, Liu MQ, Ma L, Yu SY. Volume gain of brainstem on medication-overuse headache using voxel-based morphometry. Chin Med J (Engl). 2018;131:21582163. Chen ZY, Chen XY, Liu MQ, Ma L, Yu SY. Volume gain of brainstem on medication-overuse headache using voxel-based morphometry. Chin Med J (Engl). 2018;131:21582163.
46.
go back to reference Chong CD, Dumkrieger GM, Schwedt TJ. Structural co-variance patterns in migraine: a cross-sectional study exploring the role of thehippocampus. Headache. 2017;57(10):1522–31.PubMedPubMedCentralCrossRef Chong CD, Dumkrieger GM, Schwedt TJ. Structural co-variance patterns in migraine: a cross-sectional study exploring the role of thehippocampus. Headache. 2017;57(10):1522–31.PubMedPubMedCentralCrossRef
47.
go back to reference Hubbard CS, Khan SA, Keaser ML, Mathur VA, Goyal M, Seminowicz DA. Altered brain structure and function correlate with disease severity and pain catastrophizing in migraine patients. eNeuro. 2014;1(1):e20.14.PubMedCrossRef Hubbard CS, Khan SA, Keaser ML, Mathur VA, Goyal M, Seminowicz DA. Altered brain structure and function correlate with disease severity and pain catastrophizing in migraine patients. eNeuro. 2014;1(1):e20.14.PubMedCrossRef
48.
go back to reference Liu HY, Chou KH, Lee PL, Fuh JL, Niddam DM, Lai KL, et al. Hippocampus and amygdala volume in relation to migraine frequency and prognosis. Cephalalgia. 2016;37(14):1329–36.PubMedCrossRef Liu HY, Chou KH, Lee PL, Fuh JL, Niddam DM, Lai KL, et al. Hippocampus and amygdala volume in relation to migraine frequency and prognosis. Cephalalgia. 2016;37(14):1329–36.PubMedCrossRef
49.
go back to reference Liu J, Lan L, Mu J, Zhao L, Yuan K, Zhang Y, et al. Genetic contributionofcatechol-O-methyltransferaseinhippocampalstructural and functional changes of female migraine sufferers. Hum Brain Mapp. 2015;36(5):1782–95.PubMedPubMedCentralCrossRef Liu J, Lan L, Mu J, Zhao L, Yuan K, Zhang Y, et al. Genetic contributionofcatechol-O-methyltransferaseinhippocampalstructural and functional changes of female migraine sufferers. Hum Brain Mapp. 2015;36(5):1782–95.PubMedPubMedCentralCrossRef
50.
go back to reference Maleki N, Becerra L, Brawn J, McEwen B, Burstein R, Borsook D. Common hippocampal structural and functional changes in migraine. Brain Struct Funct. 2013;218(4):903–12.PubMedCrossRef Maleki N, Becerra L, Brawn J, McEwen B, Burstein R, Borsook D. Common hippocampal structural and functional changes in migraine. Brain Struct Funct. 2013;218(4):903–12.PubMedCrossRef
51.
go back to reference Liu J, Lan L, Li G, Yan X, Nan J, Xiong S, et al. Migraine-related gray matter and white matter changes at a 1-year follow-up evaluation. J Pain. 2013;14(12):1703–8.PubMedCrossRef Liu J, Lan L, Li G, Yan X, Nan J, Xiong S, et al. Migraine-related gray matter and white matter changes at a 1-year follow-up evaluation. J Pain. 2013;14(12):1703–8.PubMedCrossRef
52.
go back to reference Coppola G, Petolicchio B, Di Renzo A, Tinelli E, Di Lorenzo C, Parisi V, et al. Cerebral gray matter volume in patients with chronic migraine: correlations with clinical features. J Headache Pain. 2017;18:115.PubMedPubMedCentralCrossRef Coppola G, Petolicchio B, Di Renzo A, Tinelli E, Di Lorenzo C, Parisi V, et al. Cerebral gray matter volume in patients with chronic migraine: correlations with clinical features. J Headache Pain. 2017;18:115.PubMedPubMedCentralCrossRef
53.
go back to reference Neeb L, Bastian K, Villringer K, Israel H, Reuter U, Fiebach JB. Structural gray matter alterations in chronic migraine: implications for a progressive disease? Headache. 2017;57:400416.CrossRef Neeb L, Bastian K, Villringer K, Israel H, Reuter U, Fiebach JB. Structural gray matter alterations in chronic migraine: implications for a progressive disease? Headache. 2017;57:400416.CrossRef
54.
go back to reference Schulte LH, Allers A, May A. Hypothalamus as a mediator of chronic migraine: evidence from high-resolution fMRI. Neurology. 2017;88:2011–6.PubMedCrossRef Schulte LH, Allers A, May A. Hypothalamus as a mediator of chronic migraine: evidence from high-resolution fMRI. Neurology. 2017;88:2011–6.PubMedCrossRef
55.
go back to reference Androulakis XM, Krebs KA, Jenkins C, Maleki N, Finkel AG, Rorden C, et al. Central executive and default mode network intranet work functional connectivity patterns in chronic migraine. J Neurol Disord. 2018;6:393.PubMedPubMedCentralCrossRef Androulakis XM, Krebs KA, Jenkins C, Maleki N, Finkel AG, Rorden C, et al. Central executive and default mode network intranet work functional connectivity patterns in chronic migraine. J Neurol Disord. 2018;6:393.PubMedPubMedCentralCrossRef
56.
go back to reference Domínguez C, López A, Ramos-Cabrer P, Vieites-Prado A, Pérez-Mato M, Villalba C, et al. Iron deposition in periaqueductal gray matter as a potential biomarker for chronic migraine. Neurology. 2019;92:e1076–85.PubMedCrossRef Domínguez C, López A, Ramos-Cabrer P, Vieites-Prado A, Pérez-Mato M, Villalba C, et al. Iron deposition in periaqueductal gray matter as a potential biomarker for chronic migraine. Neurology. 2019;92:e1076–85.PubMedCrossRef
57.
go back to reference Lee MJ, Park BY, Cho S, Kim ST, Park H, Chung CS. Increased connectivity of pain matrix in chronic migraine: a resting-state functional MRI study. J Headache Pain. 2019;20:29.PubMedPubMedCentralCrossRef Lee MJ, Park BY, Cho S, Kim ST, Park H, Chung CS. Increased connectivity of pain matrix in chronic migraine: a resting-state functional MRI study. J Headache Pain. 2019;20:29.PubMedPubMedCentralCrossRef
58.
go back to reference Woldeamanuel YW, DeSouza DD, Sanjanwala BM, Cowan RP. Clinical features contributing to cortical thickness changes in chronic migraine—a pilot study. Headache. 2019;59:180–91.PubMedCrossRef Woldeamanuel YW, DeSouza DD, Sanjanwala BM, Cowan RP. Clinical features contributing to cortical thickness changes in chronic migraine—a pilot study. Headache. 2019;59:180–91.PubMedCrossRef
59.
go back to reference Kim JH, Suh SI, Seol HY, Oh K, Seo WK, Yu SW, et al. Regional grey matter changes in patients with migraine: a voxel-based morphometry study. Cephalalgia. 2008;28:598–604.PubMedCrossRef Kim JH, Suh SI, Seol HY, Oh K, Seo WK, Yu SW, et al. Regional grey matter changes in patients with migraine: a voxel-based morphometry study. Cephalalgia. 2008;28:598–604.PubMedCrossRef
60.
go back to reference Valfrè W, Rainero I, Bergui M, Pinessi L. Voxel-based morphometry reveals gray matter abnormalities in migraine. Headache. 2008;48:109–17.PubMedCrossRef Valfrè W, Rainero I, Bergui M, Pinessi L. Voxel-based morphometry reveals gray matter abnormalities in migraine. Headache. 2008;48:109–17.PubMedCrossRef
61.
go back to reference Ong WY, Stohler CS, Herr DR. Role of the prefrontal cortex in pain processing. Mol Neurobiol. 2019;56:1137–66.PubMedCrossRef Ong WY, Stohler CS, Herr DR. Role of the prefrontal cortex in pain processing. Mol Neurobiol. 2019;56:1137–66.PubMedCrossRef
62.
go back to reference Krummenacher P, Candia V, Folkers G, Schedlowski M, Schönbächler G. Prefrontal cortex modulates placebo analgesia. Pain. 2010;148:368–74.PubMedCrossRef Krummenacher P, Candia V, Folkers G, Schedlowski M, Schönbächler G. Prefrontal cortex modulates placebo analgesia. Pain. 2010;148:368–74.PubMedCrossRef
63.
go back to reference Bräscher AK, Becker S, Hoeppli ME, Schweinhardt P. Different brain circuitries mediating controllable and uncontrollable pain. J Neurosci. 2016;36:5013–25.PubMedPubMedCentralCrossRef Bräscher AK, Becker S, Hoeppli ME, Schweinhardt P. Different brain circuitries mediating controllable and uncontrollable pain. J Neurosci. 2016;36:5013–25.PubMedPubMedCentralCrossRef
64.
65.
go back to reference May A. Experience-dependent structural plasticity in the adult human brain. Trends Cogn Sci. 2011;15:475–82.PubMedCrossRef May A. Experience-dependent structural plasticity in the adult human brain. Trends Cogn Sci. 2011;15:475–82.PubMedCrossRef
Metadata
Title
A volumetric magnetic resonance imaging study in migraine
Authors
Laila Elmously Naguib
Ghada Saed Abdel Azim
Mohammed Abdelrazek Abdellatif
Publication date
01-12-2021
Publisher
Springer Berlin Heidelberg
DOI
https://doi.org/10.1186/s41983-021-00372-7

Other articles of this Issue 1/2021

The Egyptian Journal of Neurology, Psychiatry and Neurosurgery 1/2021 Go to the issue