1
|
Liu D, Wang T, Wang Q, Dong P, Liu X, Li Q, Shi Y, Li J, Zhou J, Zhang Q. Identification of key genes in sepsis-induced cardiomyopathy based on integrated bioinformatical analysis and experiments in vitro and in vivo. PeerJ 2023; 11:e16222. [PMID: 38025678 PMCID: PMC10668858 DOI: 10.7717/peerj.16222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/11/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Sepsis is a life-threatening disease that damages multiple organs and induced by the host's dysregulated response to infection with high morbidity and mortality. Heart remains one of the most vulnerable targets of sepsis-induced organ damage, and sepsis-induced cardiomyopathy (SIC) is an important factor that exacerbates the death of patients. However, the underlying genetic mechanism of SIC disease needs further research. Methods The transcriptomic dataset, GSE171564, was downloaded from NCBI for further analysis. Gene expression matrices for the sample group were obtained by quartile standardization and log2 logarithm conversion prior to analysis. The time series, protein-protein interaction (PPI) network, and functional enrichment analysis via Gene Ontology and KEGG Pathway Databases were used to identify key gene clusters and their potential interactions. Predicted miRNA-mRNA relationships from multiple databases facilitated the construction of a TF-miRNA-mRNA regulatory network. In vivo experiments, along with qPCR and western blot assays, provided experimental validation. Results The transcriptome data analysis between SIC and healthy samples revealed 221 down-regulated, and 342 up-regulated expressed genes across two distinct clusters. Among these, Tpt1, Mmp9 and Fth1 were of particular significance. Functional analysis revealed their role in several biological processes and pathways, subsequently, in vivo experiments confirmed their overexpression in SIC samples. Notably, we found TPT1 play a pivotal role in the progression of SIC, and silencing TPT1 showed a protective effect against LPS-induced SIC. Conclusion In our study, we demonstrated that Tpt1, Mmp9 and Fth1 have great potential to be biomarker of SIC. These findings will facilitated to understand the occurrence and development mechanism of SIC.
Collapse
Affiliation(s)
- Dehua Liu
- Weifang Medical University, Weifang, China
| | - Tao Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Qingguo Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Peikang Dong
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaohong Liu
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Qiang Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Youkui Shi
- Department of Emergency Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingtian Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jin Zhou
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Quan Zhang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
2
|
Messina R, Rocca MA, Goadsby PJ, Filippi M. Insights into migraine attacks from neuroimaging. Lancet Neurol 2023; 22:834-846. [PMID: 37478888 DOI: 10.1016/s1474-4422(23)00152-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/15/2023] [Accepted: 04/12/2023] [Indexed: 07/23/2023]
Abstract
Migraine is one of the most common neurological diseases and it has a huge social and personal impact. Although head pain is the core symptom, individuals with migraine can have a plethora of non-headache symptoms that precede, accompany, or follow the pain. Neuroimaging studies have shown that the involvement of specific brain areas can explain many of the symptoms reported during the different phases of migraine. Recruitment of the hypothalamus, pons, spinal trigeminal nucleus, thalamus, and visual and pain-processing cortical areas starts during the premonitory phase and persists through the headache phase, contributing to the onset of pain and associated symptoms. Once the pain stops, the involvement of most brain areas ends, although the pons, hypothalamus, and visual cortex remain active after acute treatment intake and resolution of migraine symptoms. A better understanding of the correlations between imaging findings and migraine symptomatology can provide new insight into migraine pathophysiology and the mechanisms of novel migraine-specific treatments.
Collapse
Affiliation(s)
- Roberta Messina
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Peter J Goadsby
- NIHR King's Clinical Research Facility, King's College, London, UK; Department of Neurology, University of California, Los Angeles, CA, USA
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy; Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
3
|
Messina R, Filippi M. What imaging has revealed about migraine and chronic migraine. HANDBOOK OF CLINICAL NEUROLOGY 2023; 198:105-116. [PMID: 38043956 DOI: 10.1016/b978-0-12-823356-6.00011-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Although migraine pathophysiology is not yet entirely understood, it is now established that migraine should be viewed as a complex neurological disease, which involves the interplay of different brain networks and the release of signaling molecules, instead of a pure vascular disorder. The field of migraine research has also progressed significantly due to the advancement of brain imaging techniques. Numerous studies have investigated the relation between migraine pathophysiology and cerebral hemodynamic changes, showing that vascular changes are neither necessary nor sufficient to cause the migraine pain. Abnormal function and structure of key cortical, subcortical, and brainstem regions involved in multisensory, including pain, processing have been shown to occur in migraine patients during both an acute attack and the interictal phase. Whether brain imaging alterations represent a predisposing trait or are the consequence of the recurrence of headache attacks is still a matter of debate. It is highly likely that brain functional and structural alterations observed in migraine patients derive from the interaction between predisposing brain traits and experience-dependent responses. Neuroimaging studies have also enriched our knowledge of the mechanisms responsible for migraine chronification and have shed light on the mechanisms of actions of acute and preventive migraine treatments.
Collapse
Affiliation(s)
- Roberta Messina
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy; Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
4
|
Koehler PJ, Boes CJ. History of migraine. HANDBOOK OF CLINICAL NEUROLOGY 2023; 198:3-21. [PMID: 38043968 DOI: 10.1016/b978-0-12-823356-6.00002-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Migraine symptoms were described in ancient Babylonia, and supernatural forces were felt to play a role in etiology and treatment. This changed in the Greco-Roman period, when the (dis)balance of humors was considered in (patho)physiology and treatment based on this. Aretaeus distinguished between cephalalgia, cephalea, and heterocrania. The latter term was changed to hemicrania by Galen. Physicians in the 17th century attributed headache to the meninges, extracranial periost, and cranial blood vessels. As for the pathophysiology, Willis suggested intracranial vasoconstriction with subsequent dilatation. Tissot and Fothergill gave comprehensive descriptions of migraine, including visual symptoms. Symptomatic and idiopathic hemicrania were distinguished in the early 19th century. Vasomotor pathophysiology was scientifically studied in the 1860s, leading to sympathicotonic and angioparalytic theories. Latham combined them, stating the latter follows the first. Ergot was introduced in 1868; ergotamine was isolated in 1918. This led to the vasodilatation theory of migraine (Wolff), the discovery of 5-HT, and later the specific agonists. Aura and cortical spreading depression were studied in the early 1940s and related to spreading oligemia in the 1980s. Subsequently, hyperemia followed by oligemia after CSD was found. After the discovery of CGRP, a new a class of drugs became the subject of clinical studies.
Collapse
Affiliation(s)
- Peter J Koehler
- Faculty of Health, Medicine & Life Sciences, University of Maastricht, Maastricht, The Netherlands.
| | | |
Collapse
|
5
|
Monteith TS. Advocacy for Migraine Relief: Strategic Planning to Eliminate the Burden. Curr Pain Headache Rep 2022; 26:567-574. [PMID: 35716274 PMCID: PMC9206221 DOI: 10.1007/s11916-022-01059-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW The goal of this article is to describe migraine advocacy as an essential means to advance the field of headache medicine. Special attention is spent outlining advocacy initiatives and priorities. RECENT FINDINGS There is little written about "migraine advocacy" in the literature. However, organizational and expert priorities include advocating for policies that improve systems of care, telemedicine, education, research, and public initiatives that reduce health disparities and the stigma of migraine. This summary includes the latest advocacy efforts to support policies that may improve migraine care, strengthen the field of headache medicine, and eliminate the burden of migraine.
Collapse
|
6
|
Chen Y, Kang Y, Luo S, Liu S, Wang B, Gong Z, Huang Y, Wang H, Zhan S, Tan W. The cumulative therapeutic effect of acupuncture in patients with migraine without aura: Evidence from dynamic alterations of intrinsic brain activity and effective connectivity. Front Neurosci 2022; 16:925698. [PMID: 35928016 PMCID: PMC9344052 DOI: 10.3389/fnins.2022.925698] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
We explored the dynamic alterations of intrinsic brain activity and effective connectivity after acupuncture treatment to investigate the underlying neurological mechanism of acupuncture treatment in patients with migraine without aura (MwoA). The Functional Magnetic Resonance Imaging (fMRI) scans were separately obtained at baseline, after the first and 12th acupuncture sessions in 40 patients with MwoA. Compared with the healthy controls (HCs), patients with MwoA mostly showed a decreased dynamic amplitude of low-frequency fluctuation (dALFF) variability in the rostral ventromedial medulla (RVM), superior lobe of left cerebellum (Cerebellum_Crus1_L), right precuneus (PCUN.R), and so on. The decreased dALFF variability of RVM, Cerebellum_Crus1_L, and PCUN.R progressively recovered after the first and 12th acupuncture treatment sessions as compared to the baseline. There was gradually increased dynamic effective connectivity (DEC) variability in RVM outflow to the right middle frontal gyrus, left insula, right precentral gyrus, and right supramarginal gyrus, and gradually enhanced DEC variability from the right fusiform gyrus inflow to RVM. Furthermore, the gradually increased DEC variability was found from Cerebellum_Crus1_L outflow to the left middle occipital gyrus and the left precentral gyrus, from PCUN.R outflow to the right thalamus. These dALFF variabilities were positively correlated with the frequency of migraine attacks and negatively correlated with disease duration at baseline. The dynamic Granger causality analysis (GCA) coefficients of this DEC variability were positively correlated with Migraine-Specific Quality of Life Questionnaire scores and negatively correlated with the frequency of migraine attacks and visual analog scale (VAS) scores after 12th acupuncture sessions. Our results were analyzed by a longitudinal fMRI in the absence of a sham acupuncture control group and provided insight into the dynamic alterations of brain activity and effective connectivity in patients with MwoA after acupuncture intervention. Acupuncture might relieve MwoA by increasing the effective connectivity of RVM, Cerebellum_Crus1_L, and PCUN.R to make up for the decreased dALFF variability in these brain areas.
Collapse
Affiliation(s)
- Yilei Chen
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingjie Kang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shilei Luo
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shanshan Liu
- Department of Acupuncture and Moxibustion, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bo Wang
- Department of Acupuncture and Moxibustion, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhigang Gong
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanwen Huang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Wang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Songhua Zhan
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Songhua Zhan,
| | - Wenli Tan
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Wenli Tan,
| |
Collapse
|
7
|
Association between migraine and risk of ocular motor cranial nerve palsy. Sci Rep 2022; 12:10512. [PMID: 35732687 PMCID: PMC9217919 DOI: 10.1038/s41598-022-14621-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 06/09/2022] [Indexed: 11/20/2022] Open
Abstract
To assess association between migraines and development of ocular motor cranial nerve palsy (CNP) and finding risk factors using the National Sample Cohort database from the Korea National Health Insurance Service. Data was analyzed from 4,234,341 medical screening examinees aged 20–90 years in 2009. Cox proportional hazard regression analysis was used to the adjusted hazard ratios (HR) for ocular motor CNP according to presence of migraine. Subgroup analysis was performed to evaluate effect of other factors on association of migraine with ocular motor CNP. A total of 5806 participants (0.14% of subjects) developed ocular motor CNP and were assigned to CNP group, 4,048,018 were assigned to control group, with an average of 8.22 ± 0.93 years of follow-up. Incidence of ocular motor CNP increased in migraine group compared to control. After adjusting potential confounding variables, HR for ocular motor CNP was 1.166 (confidence interval [CI] 1.013–1.343) in migraine group. Subgroups of relatively younger age less than 65 years (HR = 1.267, 95% CI 1.067–1.504), male gender (HR = 1.228, 95% CI 1.000–1.122), smokers (HR 1.426, 95% CI 1.127–1.803), and diabetes mellitus patients (HR = 1.378, 95% CI 1.045–1.378) showed a stronger association between migraines and development of ocular motor CNP. Our population-based cohort study demonstrated a significant association between presence of migraines and incidence of ocular motor CNP. Especially, relatively younger age, males, smokers, and diabetes patients with migraines could have a higher risk of developing ocular motor CNP.
Collapse
|
8
|
Hautakangas H, Winsvold BS, Ruotsalainen SE, Bjornsdottir G, Harder AVE, Kogelman LJA, Thomas LF, Noordam R, Benner C, Gormley P, Artto V, Banasik K, Bjornsdottir A, Boomsma DI, Brumpton BM, Burgdorf KS, Buring JE, Chalmer MA, de Boer I, Dichgans M, Erikstrup C, Färkkilä M, Garbrielsen ME, Ghanbari M, Hagen K, Häppölä P, Hottenga JJ, Hrafnsdottir MG, Hveem K, Johnsen MB, Kähönen M, Kristoffersen ES, Kurth T, Lehtimäki T, Lighart L, Magnusson SH, Malik R, Pedersen OB, Pelzer N, Penninx BWJH, Ran C, Ridker PM, Rosendaal FR, Sigurdardottir GR, Skogholt AH, Sveinsson OA, Thorgeirsson TE, Ullum H, Vijfhuizen LS, Widén E, van Dijk KW, Aromaa A, Belin AC, Freilinger T, Ikram MA, Järvelin MR, Raitakari OT, Terwindt GM, Kallela M, Wessman M, Olesen J, Chasman DI, Nyholt DR, Stefánsson H, Stefansson K, van den Maagdenberg AMJM, Hansen TF, Ripatti S, Zwart JA, Palotie A, Pirinen M. Genome-wide analysis of 102,084 migraine cases identifies 123 risk loci and subtype-specific risk alleles. Nat Genet 2022; 54:152-160. [PMID: 35115687 PMCID: PMC8837554 DOI: 10.1038/s41588-021-00990-0] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 11/22/2021] [Indexed: 12/11/2022]
Abstract
Migraine affects over a billion individuals worldwide but its genetic underpinning remains largely unknown. Here, we performed a genome-wide association study of 102,084 migraine cases and 771,257 controls and identified 123 loci, of which 86 are previously unknown. These loci provide an opportunity to evaluate shared and distinct genetic components in the two main migraine subtypes: migraine with aura and migraine without aura. Stratification of the risk loci using 29,679 cases with subtype information indicated three risk variants that seem specific for migraine with aura (in HMOX2, CACNA1A and MPPED2), two that seem specific for migraine without aura (near SPINK2 and near FECH) and nine that increase susceptibility for migraine regardless of subtype. The new risk loci include genes encoding recent migraine-specific drug targets, namely calcitonin gene-related peptide (CALCA/CALCB) and serotonin 1F receptor (HTR1F). Overall, genomic annotations among migraine-associated variants were enriched in both vascular and central nervous system tissue/cell types, supporting unequivocally that neurovascular mechanisms underlie migraine pathophysiology.
Collapse
Affiliation(s)
- Heidi Hautakangas
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Bendik S Winsvold
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Sanni E Ruotsalainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | | | - Aster V E Harder
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Lisette J A Kogelman
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Laurent F Thomas
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Christian Benner
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | | | - Ville Artto
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Karina Banasik
- Novo Nordic Foundation Center for Protein Research, Copenhagen University, Copenhagen, Denmark
| | | | - Dorret I Boomsma
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | - Ben M Brumpton
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Julie E Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mona Ameri Chalmer
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Irene de Boer
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Markus Färkkilä
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Maiken Elvestad Garbrielsen
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Knut Hagen
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinical Research Unit Central Norway, St. Olavs University Hospital, Trondheim, Norway
| | - Paavo Häppölä
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Jouke-Jan Hottenga
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | | | - Kristian Hveem
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Center, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marianne Bakke Johnsen
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research and Communication Unit for Musculoskeletal Health (FORMI), Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Espen S Kristoffersen
- Research and Communication Unit for Musculoskeletal Health (FORMI), Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of General Practice, Institute of Health and Society, University of Oslo, Oslo, Norway
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Tobias Kurth
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Lannie Lighart
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | | | - Rainer Malik
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Ole Birger Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Nadine Pelzer
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
- GGZ inGeest Specialized Mental Health Care, Amsterdam, the Netherlands
| | - Caroline Ran
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Anne Heidi Skogholt
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Lisanne S Vijfhuizen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arpo Aromaa
- National Public Health Institute (Finnish Institute for Health and Welfare - THL), Helsinki, Finland
| | | | - Tobias Freilinger
- Klinikum Passau, Department of Neurology, Passau, Germany
- Centre of Neurology, Hertie Institute for Clinical Brain Research, Tuebingen, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marjo-Riitta Järvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mikko Kallela
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Maija Wessman
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Jes Olesen
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Dale R Nyholt
- School of Biomedical Sciences and Centre for Genomics and Personalised Health, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | | | | | - Arn M J M van den Maagdenberg
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas Folkmann Hansen
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Copenhagen, Denmark
- Novo Nordic Foundation Center for Protein Research, Copenhagen University, Copenhagen, Denmark
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - John-Anker Zwart
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
- Department of Public Health, University of Helsinki, Helsinki, Finland.
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
9
|
Biscetti L, De Vanna G, Cresta E, Bellotti A, Corbelli I, Letizia Cupini M, Calabresi P, Sarchielli P. Immunological findings in patients with migraine and other primary headaches: a narrative review. Clin Exp Immunol 2022; 207:11-26. [PMID: 35020858 PMCID: PMC8802184 DOI: 10.1093/cei/uxab025] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 11/14/2021] [Accepted: 11/24/2021] [Indexed: 12/17/2022] Open
Abstract
Experimental findings suggest an involvement of neuroinflammatory mechanisms in the pathophysiology of migraine. Specifically, preclinical models of migraine have emphasized the role of neuroinflammation following the activation of the trigeminal pathway at several peripheral and central sites including dural vessels, the trigeminal ganglion, and the trigeminal nucleus caudalis. The evidence of an induction of inflammatory events in migraine pathophysiological mechanisms has prompted researchers to investigate the human leukocyte antigen (HLA) phenotypes as well as cytokine genetic polymorphisms in order to verify their potential relationship with migraine risk and severity. Furthermore, the role of neuroinflammation in migraine seems to be supported by evidence of an increase in pro-inflammatory cytokines, both ictally and interictally, together with the prevalence of Th1 lymphocytes and a reduction in regulatory lymphocyte subsets in peripheral blood of migraineurs. Cytokine profiles of cluster headache (CH) patients and those of tension-type headache patients further suggest an immunological dysregulation in the pathophysiology of these primary headaches, although evidence is weaker than for migraine. The present review summarizes available findings to date from genetic and biomarker studies that have explored the role of inflammation in primary headaches.
Collapse
Affiliation(s)
- Leonardo Biscetti
- Istituto Nazionale di Riposo e Cura dell'Anziano a carattere scientifico, IRCSS-INRCA, Ancona, Italy
| | - Gioacchino De Vanna
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Cresta
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessia Bellotti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ilenia Corbelli
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Paolo Calabresi
- Department of Neuroscience, Università Cattolica Sacro Cuore, Rome, Italy.,Neurologia, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Paola Sarchielli
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
Abstract
Photophobia is one of the most common symptoms in migraine, and the underlying mechanism is uncertain. The discovery of the intrinsically-photosensitive retinal ganglion cells which signal the intensity of light on the retina has led to discussion of their role in the pathogenesis of photophobia. In the current review, we discuss the relationship between pain and discomfort leading to light aversion (traditional photophobia) and discomfort from flicker, patterns, and colour that are also common in migraine and cannot be explained solely by the activity of intrinsically-photosensitive retinal ganglion cells. We argue that, at least in migraine, a cortical mechanism provides a parsimonious explanation for discomfort from all forms of visual stimulation, and that the traditional definition of photophobia as pain in response to light may be too restrictive. Future investigation that directly compares the retinal and cortical contributions to photophobia in migraine with that in other conditions may offer better specificity in identifying biomarkers and possible mechanisms to target for treatment.
Collapse
Affiliation(s)
| | - Sarah M Haigh
- Department of Psychology and Integrative Neuroscience, University of Nevada, Reno, USA
| | - Omar A Mahroo
- Institute of Ophthalmology, University College London, London, UK and Retinal Service, Moorfields Eye Hospital, London, UK
| | | |
Collapse
|
11
|
Chamanzar A, Haigh SM, Grover P, Behrmann M. Abnormalities in cortical pattern of coherence in migraine detected using ultra high-density EEG. Brain Commun 2021; 3:fcab061. [PMID: 34258580 PMCID: PMC8269966 DOI: 10.1093/braincomms/fcab061] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/28/2021] [Indexed: 11/13/2022] Open
Abstract
Individuals with migraine generally experience photophobia and/or phonophobia during and between migraine attacks. Many different mechanisms have been postulated to explain these migraine phenomena including abnormal patterns of connectivity across the cortex. The results, however, remain contradictory and there is no clear consensus on the nature of the cortical abnormalities in migraine. Here, we uncover alterations in cortical patterns of coherence (connectivity) in interictal migraineurs during the presentation of visual and auditory stimuli and during rest. We used a high-density EEG system, with 128 customized electrode locations, to compare inter- and intra-hemispheric coherence in the interictal period from 17 individuals with migraine (12 female) and 18 age- and gender-matched healthy control subjects. During presentations of visual (vertical grating pattern) and auditory (modulated tone) stimulation which varied in temporal frequency (4 and 6 Hz), and during rest, participants performed a colour detection task at fixation. Analyses included characterizing the inter- and intra-hemisphere coherence between the scalp EEG channels over 2-s time intervals and over different frequency bands at different spatial distances and spatial clusters. Pearson's correlation coefficients were estimated at zero-lag. Repeated measures analyses-of-variance revealed that, relative to controls, migraineurs exhibited significantly (i) faster colour detection performance, (ii) lower spatial coherence of alpha-band activity, for both inter- and intra-hemisphere connections, and (iii) the reduced coherence occurred predominantly in frontal clusters during both sensory conditions, regardless of the stimulation frequency, as well as during the resting-state. The abnormal patterns of EEG coherence in interictal migraineurs during visual and auditory stimuli, as well as at rest (eyes open), may be associated with the cortical hyper-responsivity that is characteristic of abnormal sensory processing in migraineurs.
Collapse
Affiliation(s)
- Alireza Chamanzar
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Sarah M Haigh
- Department of Psychology, University of Nevada, Reno, NV 89557, USA
- Institute for Neuroscience, University of Nevada, Reno, NV 89557, USA
- Department of Psychology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Pulkit Grover
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Marlene Behrmann
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Psychology, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Edvinsson L, Haanes KA. Identifying New Antimigraine Targets: Lessons from Molecular Biology. Trends Pharmacol Sci 2021; 42:217-225. [PMID: 33495027 DOI: 10.1016/j.tips.2021.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/22/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Primary headaches are one of the most common conditions; migraine being most prevalent. Recent work on the pathophysiology of migraine suggests a mismatch in the communication or tuning of the trigeminovascular system, leading to sensitization and the release of calcitonin gene-related peptide (CGRP). In the current Opinion, we use the up-to-date molecular understanding of mechanisms behind migraine pain, to provide novel aspects on how to modify the system and for the development of future treatments; acute as well as prophylactic. We explore the distribution and the expression of neuropeptides themselves, as well as certain ion channels, and most importantly how they may act in concert as modulators of excitability of both the trigeminal C neurons and the Aδ neurons.
Collapse
Affiliation(s)
- Lars Edvinsson
- Department of Clinical Experimental Research, Copenhagen University Hospital, Rigshospitalet-Glostrup, Denmark; Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden.
| | - Kristian Agmund Haanes
- Department of Clinical Experimental Research, Copenhagen University Hospital, Rigshospitalet-Glostrup, Denmark
| |
Collapse
|
13
|
de Vries T, Villalón CM, MaassenVanDenBrink A. Pharmacological treatment of migraine: CGRP and 5-HT beyond the triptans. Pharmacol Ther 2020; 211:107528. [PMID: 32173558 DOI: 10.1016/j.pharmthera.2020.107528] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/08/2020] [Indexed: 01/08/2023]
Abstract
Migraine is a highly disabling neurovascular disorder characterized by a severe headache (associated with nausea, photophobia and/or phonophobia), and trigeminovascular system activation involving the release of calcitonin-gene related peptide (CGRP). Novel anti-migraine drugs target CGRP signaling through either stimulation of 5-HT1F receptors on trigeminovascular nerves (resulting in inhibition of CGRP release) or direct blockade of CGRP or its receptor. Lasmiditan is a highly selective 5-HT1F receptor agonist and, unlike the triptans, is devoid of vasoconstrictive properties, allowing its use in patients with cardiovascular risk. Since lasmiditan can actively penetrate the blood-brain barrier, central therapeutic as well as side effects mediated by 5-HT1F receptor activation should be further investigated. Other novel anti-migraine drugs target CGRP signaling directly. This neuropeptide can be targeted by the monoclonal antibodies eptinezumab, fremanezumab and galcanezumab, or by CGRP-neutralizing L-aptamers called Spiegelmers. The CGRP receptor can be targeted by the monoclonal antibody erenumab, or by small-molecule antagonists called gepants. Currently, rimegepant and ubrogepant have been developed for acute migraine treatment, while atogepant is studied for migraine prophylaxis. Of these drugs targeting CGRP signaling directly, eptinezumab, erenumab, fremanezumab, galcanezumab, rimegepant and ubrogepant have been approved for clinical use, while atogepant is in the last stage before approval. Although all of these drugs seem highly promising for migraine treatment, their safety should be investigated in the long-term. Moreover, the exact mechanism(s) of action of these drugs need to be elucidated further, to increase both safety and efficacy and to increase the number of responders to the different treatments, so that all migraine patients can satisfactorily be treated.
Collapse
Affiliation(s)
- Tessa de Vries
- Division of Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, PO Box 2040, 3000, CA, Rotterdam, the Netherlands
| | - Carlos M Villalón
- Deptartment de Farmacobiología, Cinvestav-Coapa, C.P. 14330 Ciudad de México, Mexico
| | - Antoinette MaassenVanDenBrink
- Division of Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, PO Box 2040, 3000, CA, Rotterdam, the Netherlands.
| |
Collapse
|
14
|
Techlo TR, Rasmussen AH, Møller PL, Bøttcher M, Winther S, Davidsson OB, Olofsson IA, Chalmer MA, Kogelman LJA, Nyegaard M, Olesen J, Hansen TF. Familial analysis reveals rare risk variants for migraine in regulatory regions. Neurogenetics 2020; 21:149-157. [PMID: 32076896 PMCID: PMC7283211 DOI: 10.1007/s10048-020-00606-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/07/2020] [Indexed: 01/08/2023]
Abstract
The most recent genome-wide association study of migraine increased the total number of known migraine risk loci to 38. Still, most of the heritability of migraine remains unexplained, and it has been suggested that rare gene dysregulatory variants play an important role in migraine etiology. Addressing the missing heritability of migraine, we aim to fine-map signals from the known migraine risk loci to regulatory mechanisms and associate these to downstream genic targets. We analyzed a large cohort of whole-genome sequenced patients from extended migraine pedigrees (1040 individuals from 155 families). We test for association between rare variants segregating in regulatory regions with migraine. The findings were replicated in an independent case-control cohort (2027 migraineurs, 1650 controls). We report an increased burden of rare variants in one CpG island and three polycomb group response elements near four migraine risk loci. We found that the association is independent of the common risk variants in the loci. The regulatory regions are suggested to affect different genes than those originally tagged by the index SNPs of the migraine loci. Families with familial clustering of migraine have an increased burden of rare variants in regulatory regions near known migraine risk loci, with effects that are independent of the variants in the loci. The possible regulatory targets suggest different genes than those originally tagged by the index SNPs of the migraine loci.
Collapse
Affiliation(s)
- Tanya Ramdal Techlo
- Danish Headache Center, Department of Neurology, Rigshospitalet, Nordstjernevej 40, DK-2600, Glostrup, Denmark
| | - Andreas Høiberg Rasmussen
- Danish Headache Center, Department of Neurology, Rigshospitalet, Nordstjernevej 40, DK-2600, Glostrup, Denmark
| | - Peter L Møller
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, Aarhus, Denmark
| | - Morten Bøttcher
- Department of Cardiology, Hospital Unit West Jutland, Herning, Denmark
| | - Simon Winther
- Department of Cardiology, Hospital Unit West Jutland, Herning, Denmark.,Department of Cardiology, Aarhus University Hospital, Skejby, Aarhus, Denmark
| | - Olafur B Davidsson
- Danish Headache Center, Department of Neurology, Rigshospitalet, Nordstjernevej 40, DK-2600, Glostrup, Denmark
| | - Isa A Olofsson
- Danish Headache Center, Department of Neurology, Rigshospitalet, Nordstjernevej 40, DK-2600, Glostrup, Denmark
| | - Mona Ameri Chalmer
- Danish Headache Center, Department of Neurology, Rigshospitalet, Nordstjernevej 40, DK-2600, Glostrup, Denmark
| | - Lisette J A Kogelman
- Danish Headache Center, Department of Neurology, Rigshospitalet, Nordstjernevej 40, DK-2600, Glostrup, Denmark
| | - Mette Nyegaard
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, Aarhus, Denmark
| | - Jes Olesen
- Danish Headache Center, Department of Neurology, Rigshospitalet, Nordstjernevej 40, DK-2600, Glostrup, Denmark
| | - Thomas Folkmann Hansen
- Danish Headache Center, Department of Neurology, Rigshospitalet, Nordstjernevej 40, DK-2600, Glostrup, Denmark. .,Institute for Biological Psychiatry, Mental Health Center Sct. Hans, Roskilde, Denmark. .,Novo Nordic Foundation Centre for Protein Research, Copenhagen University, Copenhagen, Denmark.
| |
Collapse
|
15
|
Rainero I, Vacca A, Govone F, Gai A, Pinessi L, Rubino E. Migraine: Genetic Variants and Clinical Phenotypes. Curr Med Chem 2019; 26:6207-6221. [DOI: 10.2174/0929867325666180719120215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/21/2018] [Accepted: 06/26/2018] [Indexed: 12/16/2022]
Abstract
Migraine is a common, chronic neurovascular disorder caused by a complex interaction
between genetic and environmental risk factors. In the last two decades, molecular genetics
of migraine have been intensively investigated. In a few cases, migraine is transmitted as a
monogenic disorder, and the disease phenotype cosegregates with mutations in different genes
like CACNA1A, ATP1A2, SCN1A, KCNK18, and NOTCH3. In the common forms of migraine,
candidate genes as well as genome-wide association studies have shown that a large number of
genetic variants may increase the risk of developing migraine. At present, few studies investigated
the genotype-phenotype correlation in patients with migraine. The purpose of this review
was to discuss recent studies investigating the relationship between different genetic variants
and the clinical characteristics of migraine. Analysis of genotype-phenotype correlations in
migraineurs is complicated by several confounding factors and, to date, only polymorphisms
of the MTHFR gene have been shown to have an effect on migraine phenotype. Additional
genomic studies and network analyses are needed to clarify the complex pathways underlying
migraine and its clinical phenotypes.
Collapse
Affiliation(s)
- Innocenzo Rainero
- Headache Center, Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Torino, Italy
| | - Alessandro Vacca
- Headache Center, Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Torino, Italy
| | - Flora Govone
- Headache Center, Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Torino, Italy
| | - Annalisa Gai
- Headache Center, Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Torino, Italy
| | - Lorenzo Pinessi
- Headache Center, Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Torino, Italy
| | - Elisa Rubino
- Headache Center, Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Torino, Italy
| |
Collapse
|
16
|
Lombardo SD, Mazzon E, Basile MS, Cavalli E, Bramanti P, Nania R, Fagone P, Nicoletti F, Petralia MC. Upregulation of IL-1 Receptor Antagonist in a Mouse Model of Migraine. Brain Sci 2019; 9:E172. [PMID: 31331109 PMCID: PMC6680509 DOI: 10.3390/brainsci9070172] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023] Open
Abstract
Migraine is a disorder characterized by attacks of monolateral headaches, often accompanied by nausea, vomiting, and photophobia. Around 30% of patients also report aura symptoms. The cause of the aura is believed to be related to the cortical spreading depression (CSD), a wave of neuronal and glial depolarization originating in the occipital cortex, followed by temporary neuronal silencing. During a migraine attack, increased expression of inflammatory mediators, along with a decrease in the expression of anti-inflammatory genes, have been observed. The aim of this study was to evaluate the expression of inflammatory genes, in particular that of IL-1 receptor antagonist (IL-1RN), following CSD in a mouse model of familial hemiplegic migraine type 1 (FHM-1). We show here that the expression of IL-1RN was upregulated after the CSD, suggesting a possible attempt to modulate the inflammatory response. This study allows researchers to better understand the development of the disease and aids in the search for new therapeutic strategies in migraine.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Eugenio Cavalli
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy
| | - Riccardo Nania
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy.
| | - Maria Cristina Petralia
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy
| |
Collapse
|
17
|
Abstract
Migraine is a strongly disabling disease characterized by a unilateral throbbing headache lasting for up to 72 h for each individual attack. There have been many theories on the pathophysiology of migraine throughout the years. Currently, the neurovascular theory dominates, suggesting clear involvement of the trigeminovascular system. The most recent data show that a migraine attack most likely originates in the hypothalamus and activates the trigeminal nucleus caudalis (TNC). Although the mechanisms are unknown, activation of the TNC leads to peripheral release of calcitonin gene-related protein (CGRP), most likely from C-fibers. During the past year monoclonal antibodies against CGRP or the CGRP receptor have emerged as the most promising targets for migraine therapy, and at the same time established the strong involvement of CGRP in the pathophysiology of migraine. The viewpoint presented here focuses further on the activation of the CGRP receptor on the sensory Aδ-fiber, leading to the sensation of pain. The CGRP receptor activates adenylate cyclase, which leads to an increase in cyclic adenosine monophosphate (cAMP). We hypothesize that cAMP activates the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, triggering an action potential sensed as pain. The mechanisms behind migraine pain on a molecular level, particularly their importance to cAMP, provide clues to potential new anti-migraine targets. In this article we focus on the development of targets related to the CGRP system, and further include novel targets such as the pituitary adenylate cyclase-activating peptide (PACAP) system, the serotonin 5-HT1F receptor, purinergic receptors, HCN channels, adenosine triphosphate-sensitive potassium channels (KATP), and the glutaminergic system.
Collapse
|
18
|
Harriott AM, Takizawa T, Chung DY, Chen SP. Spreading depression as a preclinical model of migraine. J Headache Pain 2019; 20:45. [PMID: 31046659 PMCID: PMC6734429 DOI: 10.1186/s10194-019-1001-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/18/2019] [Indexed: 01/12/2023] Open
Abstract
Spreading depression (SD) is a slowly propagating wave of near-complete depolarization of neurons and glial cells across the cortex. SD is thought to contribute to the underlying pathophysiology of migraine aura, and possibly also an intrinsic brain activity causing migraine headache. Experimental models of SD have recapitulated multiple migraine-related phenomena and are considered highly translational. In this review, we summarize conventional and novel methods to trigger SD, with specific focus on optogenetic methods. We outline physiological triggers that might affect SD susceptibility, review a multitude of physiological, biochemical, and behavioral consequences of SD, and elaborate their relevance to migraine pathophysiology. The possibility of constructing a recurrent episodic or chronic migraine model using SD is also discussed.
Collapse
Affiliation(s)
- Andrea M Harriott
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Tsubasa Takizawa
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - David Y Chung
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Shih-Pin Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan. .,Brain Research Center, National Yang-Ming University, Taipei, Taiwan. .,Division of Translational Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan. .,Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
19
|
Xu R, Xiao Y, Liu Y, Wang B, Li X, Huo C, Jia X, Hou L, Wang X. Fluorescence-Based High Throughput Screening Technologies for Natural Chloride Ion Channel Blockers. Chem Res Toxicol 2018; 31:1332-1338. [PMID: 30456946 DOI: 10.1021/acs.chemrestox.8b00205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chloride channels represent a group of potential drug targets; their blockers showed significant protecting effect on impaired cells by modulating apoptosis, autophagy, and other cell signals. However, clinical drugs with chloride channel inhibitory properties have not yet been developed. Natural product extract becomes an underlying candidate satisfied the clinical requirements for its low toxicity, low cost, and abundant sources. Here, a fluorescence-based EYFP-H148Q/I153L-HeLa cell line model was constructed by molecular cloning and verified by real-time polymerase chain reaction and Western blotting assay. By using this chloride channel blocker screening model, seven hit compounds chosen from 6988 natural compounds showed the channel blocking activity. Then the hit compounds were further validated by electrophysiological patch-clamp analysis. Our study preliminarily identified PC-4 as a new chloride channel inhibitor and demonstrated the reliability and sensitivity of fluorescence-based high throughput screening technologies for discovery of biologically active compounds from natural herbal compounds.
Collapse
Affiliation(s)
| | - Yuan Xiao
- Hong-Hui Hospital, College of Medicine , Xi'an Jiaotong University , Xi'an , China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Orr SL, Richer L, Barrowman N, Zemek R. Oral dexamethasone for the prevention of acute migraine recurrence in pediatric patients presenting to the emergency department with migraine. CEPHALALGIA REPORTS 2018. [DOI: 10.1177/2515816318804158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Objective: To assess the feasibility of a randomized controlled trial protocol that aims to determine the efficacy and safety of oral dexamethasone compared to placebo for the prevention of migraine recurrence in children and adolescents visiting the pediatric emergency department (ED) with migraine. Methods: This study was a two-arm, parallel-group, randomized, placebo-controlled, double-blind pilot trial of patients presenting to the pediatric ED with migraine. Eligible participants were randomized at 1:1 ratio to receive either oral dexamethasone 0.6 mg/kg (maximum 15 mg) or matched placebo as a single dose. Efficacy and safety outcomes were assessed at discharge, 48 h and 7 days after discharge. The primary outcome of the trial was feasibility and was assessed through participant recruitment rate, follow-up completion rates, participant satisfaction ratings and comparison of enrolled versus non-enrolled participants. Efficacy and safety outcomes were not analyzed given that this was a pilot study. Results: Twelve participants were enrolled over the 6-month recruitment period. This represents 60% of the planned sample size and a 10.5% recruitment rate. No other feasibility issues were identified and patients expressed high satisfaction rates with their treatment: 90.9% were satisfied with their treatment at discharge and at 48-h follow-up and 81.8% were satisfied with their treatment at 7-day follow-up (81.8%). There were no significant differences observed when comparing enrolled participants to those not enrolled. Conclusion: This pilot randomized controlled trial is the first to assess dexamethasone in the pediatric ED for the prevention of migraine recurrence. The protocol is feasible but recruitment in a single center was lower than expected. Future pediatric ED migraine studies may use innovative or pragmatic trial designs to maximize feasibility from a recruitment standpoint.
Collapse
Affiliation(s)
- Serena L Orr
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lawrence Richer
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, Edmonton, Alberta, Canada
| | - Nick Barrowman
- Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Roger Zemek
- Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
21
|
Ferroni P, Barbanti P, Della-Morte D, Palmirotta R, Jirillo E, Guadagni F. Redox Mechanisms in Migraine: Novel Therapeutics and Dietary Interventions. Antioxid Redox Signal 2018; 28:1144-1183. [PMID: 28990418 DOI: 10.1089/ars.2017.7260] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Migraine represents the third most prevalent and the seventh most disabling human disorder. Approximately 30% of migraine patients experience transient, fully reversible, focal neurological symptoms (aura) preceding the attack. Recent Advances: Awareness of the hypothesis that migraine actually embodies a spectrum of illnesses-ranging from episodic to chronic forms-is progressively increasing and poses novel challenges for clarifying the underlying pathophysiological mechanisms of migraine as well as for the development of novel therapeutic interventions. Several theories have evolved to the current concept that a combination of genetic, epigenetic, and environmental factors may play a role in migraine pathogenesis, although their relative importance is still being debated. CRITICAL ISSUES One critical issue that deserves a particular attention is the role of oxidative stress in migraine. Indeed, potentially harmful oxidative events occur during the migraine attack and long-lasting or frequent migraine episodes may increase brain exposure to oxidative events that can lead to chronic transformation. Moreover, a wide variety of dietary, environmental, physiological, behavioral, and pharmacological migraine triggers may act through oxidative stress, with clear implications for migraine treatment and prophylaxis. Interestingly, almost all current prophylactic migraine agents exert antioxidant effects. FUTURE DIRECTIONS Increasing awareness of the role of oxidative stress and/or decreased antioxidant defenses in migraine pathogenesis and progression to a chronic condition lays the foundations for the design of novel prophylactic approaches, which, by reducing brain oxidative phenomena, could favorably modify the clinical course of migraine. Antioxid. Redox Signal. 28, 1144-1183.
Collapse
Affiliation(s)
- Patrizia Ferroni
- 1 Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University , Rome, Italy .,2 IRCCS San Raffaele Pisana , Rome, Italy
| | - Piero Barbanti
- 3 Headache and Pain Unit, Department of Neurological, Motor and Sensorial Sciences, IRCCS San Raffaele Pisana , Rome, Italy
| | - David Della-Morte
- 1 Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University , Rome, Italy .,2 IRCCS San Raffaele Pisana , Rome, Italy .,4 Department of Systems Medicine, University of Rome "Tor Vergata ," Rome, Italy
| | - Raffaele Palmirotta
- 5 Department of Biomedical Sciences and Human Oncology, "A. Moro" University , Bari, Italy
| | - Emilio Jirillo
- 6 Department of Basic Medical Sciences, Neuroscience and Sensory Organs, "A. Moro" University , Bari, Italy
| | - Fiorella Guadagni
- 1 Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University , Rome, Italy .,2 IRCCS San Raffaele Pisana , Rome, Italy
| |
Collapse
|
22
|
Finucane HK, Reshef YA, Anttila V, Slowikowski K, Gusev A, Byrnes A, Gazal S, Loh PR, Lareau C, Shoresh N, Genovese G, Saunders A, Macosko E, Pollack S, Perry JRB, Buenrostro JD, Bernstein BE, Raychaudhuri S, McCarroll S, Neale BM, Price AL. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat Genet 2018; 50:621-629. [PMID: 29632380 PMCID: PMC5896795 DOI: 10.1038/s41588-018-0081-4] [Citation(s) in RCA: 632] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/29/2018] [Indexed: 02/07/2023]
Abstract
We introduce an approach to identify disease-relevant tissues and cell types by analyzing gene expression data together with genome-wide association study (GWAS) summary statistics. Our approach uses stratified linkage disequilibrium (LD) score regression to test whether disease heritability is enriched in regions surrounding genes with the highest specific expression in a given tissue. We applied our approach to gene expression data from several sources together with GWAS summary statistics for 48 diseases and traits (average N = 169,331) and found significant tissue-specific enrichments (false discovery rate (FDR) < 5%) for 34 traits. In our analysis of multiple tissues, we detected a broad range of enrichments that recapitulated known biology. In our brain-specific analysis, significant enrichments included an enrichment of inhibitory over excitatory neurons for bipolar disorder, and excitatory over inhibitory neurons for schizophrenia and body mass index. Our results demonstrate that our polygenic approach is a powerful way to leverage gene expression data for interpreting GWAS signals.
Collapse
Affiliation(s)
- Hilary K Finucane
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | - Yakir A Reshef
- Department of Computer Science, Harvard University, Cambridge, MA, USA
| | - Verneri Anttila
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kamil Slowikowski
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Bioinformatics and Integrative Genomics, Harvard University, Cambridge, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander Gusev
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Andrea Byrnes
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Steven Gazal
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Po-Ru Loh
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Caleb Lareau
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Noam Shoresh
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Arpiar Saunders
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Evan Macosko
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Samuela Pollack
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - John R B Perry
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - Jason D Buenrostro
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Bradley E Bernstein
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Soumya Raychaudhuri
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Partners Center for Personalized Genetic Medicine, Boston, MA, USA
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - Steven McCarroll
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Benjamin M Neale
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Alkes L Price
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
23
|
|
24
|
Okragly AJ, Morin SM, DeRosa D, Martin AP, Johnson KW, Johnson MP, Benschop RJ. Human mast cells release the migraine-inducing factor pituitary adenylate cyclase-activating polypeptide (PACAP). Cephalalgia 2017; 38:1564-1574. [PMID: 29103295 DOI: 10.1177/0333102417740563] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Many patients with migraines suffer from allergies and vice versa, suggesting a relationship between biological mechanisms of allergy and migraine. It was proposed many years ago that mast cells may be involved in the pathophysiology of migraines. We set out to investigate the relationship between mast cell activation and known neurogenic peptides related to migraine. Methods Cultured human mast cells were assayed for the presence of neuropeptides and their receptors at the RNA and protein level. Immunohistochemistry analyses were performed on tissue resident and cultured mast cells. Mast cell degranulation assays were performed and pituitary adenylate cyclase-activating polypeptide (PACAP) activity was measured with a bioassay. Results We found that cultured and tissue resident human mast cells contain PACAP in cytoplasmic granules. No other neurogenic peptide known to be involved in migraine was detected, nor did mast cells express the receptors for PACAP or other neurogenic peptides. Furthermore, mast cell degranulation through classic IgE-mediated allergic mechanisms led to the release of PACAP. The PACAP released from mast cells was biologically active, as demonstrated using PACAP receptor reporter cell lines. We confirmed existing literature that mast cell degranulation can also be induced by several neurogenic peptides, which also resulted in PACAP release. Conclusion Our data provides a potential biological explanation for the association between allergy and migraine by demonstrating the release of biologically active PACAP from mast cells.
Collapse
Affiliation(s)
- Angela J Okragly
- 1 Immunology Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - S Michelle Morin
- 2 Neuroscience Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - David DeRosa
- 1 Immunology Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Andrea P Martin
- 1 Immunology Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Kirk W Johnson
- 2 Neuroscience Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Michael P Johnson
- 2 Neuroscience Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Robert J Benschop
- 1 Immunology Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
25
|
Jay GW, Barkin RL. Primary Headache Disorders Part I- Migraine and the Trigeminal Autonomic Cephalalgias. Dis Mon 2017; 63:308-338. [DOI: 10.1016/j.disamonth.2017.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
26
|
Understanding a role for hypoxia in lesion formation and location in the deep and periventricular white matter in small vessel disease and multiple sclerosis. Clin Sci (Lond) 2017; 131:2503-2524. [PMID: 29026001 DOI: 10.1042/cs20170981] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 08/01/2017] [Accepted: 08/15/2017] [Indexed: 12/28/2022]
Abstract
The deep and periventricular white matter is preferentially affected in several neurological disorders, including cerebral small vessel disease (SVD) and multiple sclerosis (MS), suggesting that common pathogenic mechanisms may be involved in this injury. Here we consider the potential pathogenic role of tissue hypoxia in lesion development, arising partly from the vascular anatomy of the affected white matter. Specifically, these regions are supplied by a sparse vasculature fed by long, narrow end arteries/arterioles that are vulnerable to oxygen desaturation if perfusion is reduced (as in SVD, MS and diabetes) or if the surrounding tissue is hypoxic (as in MS, at least). The oxygen crisis is exacerbated by a local preponderance of veins, as these can become highly desaturated 'sinks' for oxygen that deplete it from surrounding tissues. Additional haemodynamic deficiencies, including sluggish flow and impaired vasomotor reactivity and vessel compliance, further exacerbate oxygen insufficiency. The cells most vulnerable to hypoxic damage, including oligodendrocytes, die first, resulting in demyelination. Indeed, in preclinical models, demyelination is prevented if adequate oxygenation is maintained by raising inspired oxygen concentrations. In agreement with this interpretation, there is a predilection of lesions for the anterior and occipital horns of the lateral ventricles, namely regions located at arterial watersheds, or border zones, known to be especially susceptible to hypoperfusion and hypoxia. Finally, mitochondrial dysfunction due to genetic causes, as occurs in leucodystrophies or due to free radical damage, as occurs in MS, will compound any energy insufficiency resulting from hypoxia. Viewing lesion formation from the standpoint of tissue oxygenation not only reveals that lesion distribution is partly predictable, but may also inform new therapeutic strategies.
Collapse
|
27
|
Sandor C, Beer NL, Webber C. Diverse type 2 diabetes genetic risk factors functionally converge in a phenotype-focused gene network. PLoS Comput Biol 2017; 13:e1005816. [PMID: 29059180 PMCID: PMC5667928 DOI: 10.1371/journal.pcbi.1005816] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 11/02/2017] [Accepted: 10/11/2017] [Indexed: 12/14/2022] Open
Abstract
Type 2 Diabetes (T2D) constitutes a global health burden. Efforts to uncover predisposing genetic variation have been considerable, yet detailed knowledge of the underlying pathogenesis remains poor. Here, we constructed a T2D phenotypic-linkage network (T2D-PLN), by integrating diverse gene functional information that highlight genes, which when disrupted in mice, elicit similar T2D-relevant phenotypes. Sensitising the network to T2D-relevant phenotypes enabled significant functional convergence to be detected between genes implicated in monogenic or syndromic diabetes and genes lying within genomic regions associated with T2D common risk. We extended these analyses to a recent multiethnic T2D case-control exome of 12,940 individuals that found no evidence of T2D risk association for rare frequency variants outside of previously known T2D risk loci. Examining associations involving protein-truncating variants (PTV), most at low population frequencies, the T2D-PLN was able to identify a convergent set of biological pathways that were perturbed within four of five independent T2D case/control ethnic sets of 2000 to 5000 exomes each. These same pathways were found to be over-represented among both known monogenic or syndromic diabetes genes and genes within T2D-associated common risk loci. Our study demonstrates convergent biology amongst variants representing different classes of T2D genetic risk. Although convergence was observed at the pathway level, few of the contributing genes were found in common between different cohorts or variant classes, most notably between the exome variant sets which suggests that future rare variant studies may be better focusing their power onto a single population of recent common ancestry.
Collapse
Affiliation(s)
- Cynthia Sandor
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Nicola L. Beer
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Caleb Webber
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
28
|
Robert C, Wilson CS, Lipton RB, Arreto CD. Growth of Headache Research: A 1983-2014 bibliometric study. Cephalalgia 2016; 37:1299-1309. [PMID: 27837174 DOI: 10.1177/0333102416678636] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aim This study charts the growth of the scientific journal literature on headache for 30+ years (1983-2014). Methods Using the Web of Science, articles published in four two-year periods (1983-1984, 1993-1994, 2003-2004, 2013-2014) from journals indexed in the Science Citation Index Expanded database were retrieved and analyzed. Results From 1983-1984 to 2013-2014, the scientific literature on headache increased nearly fourfold (3.8) from 468 to 1776 articles; the number of participating countries more than doubled (26 to 67); and the two most prolific countries in each period were the USA and Italy. While several European countries (Italy, Germany, UK and Denmark) were among the top 10 in each period, the notable appearance of Turkey, China and Brazil among the top 10 in 2013-2014 indicates the growing geographic spread of publications on headache research. Meanwhile, the comet-like distribution of journals has not changed: two journals, Headache and Cephalalgia, persist throughout as the nucleus, with the tail increasing more than threefold from 141 journals in 1983-1984 to 462 in 2013-2014. Conclusion Our study follows the recent growth and spread of the scientific literature on headache research and should stimulate further bibliometric investigation in this field.
Collapse
Affiliation(s)
- Claude Robert
- 1 Université Paris Descartes, Paris, France.,2 Gliaxone, Saint Germain Sous Doue, France
| | - Concepción S Wilson
- 3 School of Information Systems & Technology Management, University of New South Wales, Sydney, Australia
| | - Richard B Lipton
- 4 Department of Neurology and the Montefiore Headache Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Charles-Daniel Arreto
- 5 Université Paris Descartes, Faculté de Chirurgie Dentaire, Hôpital Bretonneau, HUPNVS, AP-HP, Paris, France
| |
Collapse
|
29
|
van Dongen RM, Zielman R, Noga M, Dekkers OM, Hankemeier T, van den Maagdenberg AM, Terwindt GM, Ferrari MD. Migraine biomarkers in cerebrospinal fluid: A systematic review and meta-analysis. Cephalalgia 2016; 37:49-63. [PMID: 26888294 DOI: 10.1177/0333102415625614] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objective To perform a meta-analysis of migraine biomarkers in cerebrospinal fluid (CSF) and of corresponding blood concentrations. Methods We conducted a systematic search for studies that measured biochemical compounds in CSF of chronic or episodic migraineurs and non-headache controls. Subsequent searches retrieved studies with blood measurements of selected CSF biomarkers. If a compound was assessed in three or more studies, results were pooled in a meta-analysis with standardised mean differences (SMD) as effect measures. Results Sixty-two compounds were measured in 40 CSF studies. Most important results include: increased glutamate (five studies, SMD 2.22, 95% CI: 1.30, 3.13), calcitonin gene-related peptide (CGRP) (three studies, SMD: 3.80, 95% CI: 3.19, 4.41) and nerve growth factor (NGF) (three studies, SMD: 6.47, 95% CI: 5.55, 7.39) in chronic migraine patients and decreased β-endorphin (β-EP) in both chronic (four studies, SMD: -1.37, 95% CI: -1.80, -0.94) and interictal episodic migraine patients (three studies, SMD: -1.12, 95% CI: -1.65, -0.58). In blood, glutamate (interictal) and CGRP (chronic, interictal and ictal) were increased and β-EP (chronic, interictal and ictal) was decreased. Conclusions Glutamate, β-EP, CGRP and NGF concentrations are altered in CSF and, except for NGF, also in blood of migraineurs. Future research should focus on the pathophysiological roles of these compounds in migraine.
Collapse
Affiliation(s)
- Robin M van Dongen
- 1 Department of Neurology, Leiden University Medical Centre, the Netherlands
| | - Ronald Zielman
- 1 Department of Neurology, Leiden University Medical Centre, the Netherlands
| | - Marek Noga
- 2 Division of Analytical Biosciences, Leiden Academic Centre for Drug Research, the Netherlands
| | - Olaf M Dekkers
- 3 Department of Clinical Epidemiology, Leiden University Medical Centre, the Netherlands.,4 Department of Clinical Epidemiology, Aarhus University Hospital, Denmark
| | - Thomas Hankemeier
- 2 Division of Analytical Biosciences, Leiden Academic Centre for Drug Research, the Netherlands
| | - Arn Mjm van den Maagdenberg
- 1 Department of Neurology, Leiden University Medical Centre, the Netherlands.,5 Department of Human Genetics, Leiden University Medical Centre, the Netherlands
| | - Gisela M Terwindt
- 1 Department of Neurology, Leiden University Medical Centre, the Netherlands
| | - Michel D Ferrari
- 1 Department of Neurology, Leiden University Medical Centre, the Netherlands
| |
Collapse
|
30
|
Gormley P, Winsvold BS, Nyholt DR, Kallela M, Chasman DI, Palotie A. Migraine genetics: from genome-wide association studies to translational insights. Genome Med 2016; 8:86. [PMID: 27543003 PMCID: PMC4992240 DOI: 10.1186/s13073-016-0346-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Padhraig Gormley
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA. .,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA. .,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| | - Bendik S Winsvold
- FORMI, Oslo University Hospital, PO 4956, Nydalen, 0424, Oslo, Norway.,Department of Neurology, Oslo University Hospital, PO 4956, Nydalen, 0424, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, PO 1171, Blindern, 0318, Oslo, Norway
| | - Dale R Nyholt
- Statistical and Genomic Epidemiology Laboratory, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Mikko Kallela
- Department of Neurology, Helsinki University Central Hospital, Haartmaninkatu 4, 00290, Helsinki, Finland
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA.,Harvard Medical School, Boston, MA, 02115, USA
| | - Aarno Palotie
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland.,Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| |
Collapse
|
31
|
Venous Thoracic Outlet Syndrome as a Cause of Intractable Migraines. Ann Vasc Surg 2016; 39:285.e5-285.e8. [PMID: 27531080 DOI: 10.1016/j.avsg.2016.05.109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 11/22/2022]
Abstract
Thoracic outlet syndrome (TOS) refers to the compression of the neurovascular bundle within the thoracic outlet. Cases are classified by primary etiology-arterial, neurogenic, or venous. In addition to the typical symptoms of arm swelling and paresthesias, headaches have been reported as a potential symptom of TOS. In this report, we describe a patient with debilitating migraines, which were consistently preceded by unilateral arm swelling. Resolution of symptoms occurred only after thoracic outlet decompression. Patients with migraines and concomitant swelling and/or paresthesias, especially related to provocative arm maneuvers, should be considered a possible atypical presentation of TOS and evaluated in more detail.
Collapse
|
32
|
Gormley P, Anttila V, Winsvold BS, Palta P, Esko T, Pers TH, Farh KH, Cuenca-Leon E, Muona M, Furlotte NA, Kurth T, Ingason A, McMahon G, Ligthart L, Terwindt GM, Kallela M, Freilinger TM, Ran C, Gordon SG, Stam AH, Steinberg S, Borck G, Koiranen M, Quaye L, Adams HHH, Lehtimäki T, Sarin AP, Wedenoja J, Hinds DA, Buring JE, Schürks M, Ridker PM, Hrafnsdottir MG, Stefansson H, Ring SM, Hottenga JJ, Penninx BWJH, Färkkilä M, Artto V, Kaunisto M, Vepsäläinen S, Malik R, Heath AC, Madden PAF, Martin NG, Montgomery GW, Kurki MI, Kals M, Mägi R, Pärn K, Hämäläinen E, Huang H, Byrnes AE, Franke L, Huang J, Stergiakouli E, Lee PH, Sandor C, Webber C, Cader Z, Muller-Myhsok B, Schreiber S, Meitinger T, Eriksson JG, Salomaa V, Heikkilä K, Loehrer E, Uitterlinden AG, Hofman A, van Duijn CM, Cherkas L, Pedersen LM, Stubhaug A, Nielsen CS, Männikkö M, Mihailov E, Milani L, Göbel H, Esserlind AL, Christensen AF, Hansen TF, Werge T, Kaprio J, Aromaa AJ, Raitakari O, Ikram MA, Spector T, Järvelin MR, Metspalu A, Kubisch C, Strachan DP, Ferrari MD, Belin AC, Dichgans M, Wessman M, van den Maagdenberg AMJM, Zwart JA, Boomsma DI, Smith GD, Stefansson K, et alGormley P, Anttila V, Winsvold BS, Palta P, Esko T, Pers TH, Farh KH, Cuenca-Leon E, Muona M, Furlotte NA, Kurth T, Ingason A, McMahon G, Ligthart L, Terwindt GM, Kallela M, Freilinger TM, Ran C, Gordon SG, Stam AH, Steinberg S, Borck G, Koiranen M, Quaye L, Adams HHH, Lehtimäki T, Sarin AP, Wedenoja J, Hinds DA, Buring JE, Schürks M, Ridker PM, Hrafnsdottir MG, Stefansson H, Ring SM, Hottenga JJ, Penninx BWJH, Färkkilä M, Artto V, Kaunisto M, Vepsäläinen S, Malik R, Heath AC, Madden PAF, Martin NG, Montgomery GW, Kurki MI, Kals M, Mägi R, Pärn K, Hämäläinen E, Huang H, Byrnes AE, Franke L, Huang J, Stergiakouli E, Lee PH, Sandor C, Webber C, Cader Z, Muller-Myhsok B, Schreiber S, Meitinger T, Eriksson JG, Salomaa V, Heikkilä K, Loehrer E, Uitterlinden AG, Hofman A, van Duijn CM, Cherkas L, Pedersen LM, Stubhaug A, Nielsen CS, Männikkö M, Mihailov E, Milani L, Göbel H, Esserlind AL, Christensen AF, Hansen TF, Werge T, Kaprio J, Aromaa AJ, Raitakari O, Ikram MA, Spector T, Järvelin MR, Metspalu A, Kubisch C, Strachan DP, Ferrari MD, Belin AC, Dichgans M, Wessman M, van den Maagdenberg AMJM, Zwart JA, Boomsma DI, Smith GD, Stefansson K, Eriksson N, Daly MJ, Neale BM, Olesen J, Chasman DI, Nyholt DR, Palotie A. Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat Genet 2016; 48:856-66. [PMID: 27322543 DOI: 10.1038/ng.3598] [Show More Authors] [Citation(s) in RCA: 454] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 05/26/2016] [Indexed: 12/16/2022]
Abstract
Migraine is a debilitating neurological disorder affecting around one in seven people worldwide, but its molecular mechanisms remain poorly understood. There is some debate about whether migraine is a disease of vascular dysfunction or a result of neuronal dysfunction with secondary vascular changes. Genome-wide association (GWA) studies have thus far identified 13 independent loci associated with migraine. To identify new susceptibility loci, we carried out a genetic study of migraine on 59,674 affected subjects and 316,078 controls from 22 GWA studies. We identified 44 independent single-nucleotide polymorphisms (SNPs) significantly associated with migraine risk (P < 5 × 10(-8)) that mapped to 38 distinct genomic loci, including 28 loci not previously reported and a locus that to our knowledge is the first to be identified on chromosome X. In subsequent computational analyses, the identified loci showed enrichment for genes expressed in vascular and smooth muscle tissues, consistent with a predominant theory of migraine that highlights vascular etiologies.
Collapse
Affiliation(s)
- Padhraig Gormley
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Verneri Anttila
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Bendik S Winsvold
- FORMI, Oslo University Hospital, Oslo, Norway.,Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Priit Palta
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Tonu Esko
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Estonian Genome Center, University of Tartu, Tartu, Estonia.,Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Tune H Pers
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Kai-How Farh
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Illumina, San Diego, California, USA
| | - Ester Cuenca-Leon
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Pediatric Neurology, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Mikko Muona
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland.,Molecular Neurology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | | | - Tobias Kurth
- Institute of Public Health, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - George McMahon
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Lannie Ligthart
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mikko Kallela
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Tobias M Freilinger
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Caroline Ran
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Scott G Gordon
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anine H Stam
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Guntram Borck
- Institute of Human Genetics, Ulm University, Ulm, Germany
| | - Markku Koiranen
- Center for Life Course Epidemiology and Systems Medicine, University of Oulu, Oulu, Finland
| | - Lydia Quaye
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, School of Medicine, University of Tampere, Tampere, Finland
| | - Antti-Pekka Sarin
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Juho Wedenoja
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | | | - Julie E Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Markus Schürks
- Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Susan M Ring
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, VU University Medical Centre, Amsterdam, the Netherlands
| | - Markus Färkkilä
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Ville Artto
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Mari Kaunisto
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Salli Vepsäläinen
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Rainer Malik
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Andrew C Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pamela A F Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicholas G Martin
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Grant W Montgomery
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mitja I Kurki
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Neurosurgery, NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Mart Kals
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Kalle Pärn
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Eija Hämäläinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Hailiang Huang
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea E Byrnes
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jie Huang
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Evie Stergiakouli
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Phil H Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Cynthia Sandor
- MRC Functional Genomics Unit, Department of Physiology, Anatomy &Genetics, Oxford University, Oxford, UK
| | - Caleb Webber
- MRC Functional Genomics Unit, Department of Physiology, Anatomy &Genetics, Oxford University, Oxford, UK
| | - Zameel Cader
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK.,Oxford Headache Centre, John Radcliffe Hospital, Oxford, UK
| | - Bertram Muller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian Albrechts University, Kiel, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,National Institute for Health and Welfare, Helsinki, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Kauko Heikkilä
- Institute of Clinical Medicine, University of Helsinki, Helsinki, Finland
| | - Elizabeth Loehrer
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lynn Cherkas
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | | | - Audun Stubhaug
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway.,Medical Faculty, University of Oslo, Oslo, Norway
| | - Christopher S Nielsen
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway.,Department of Ageing and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Minna Männikkö
- Center for Life Course Epidemiology and Systems Medicine, University of Oulu, Oulu, Finland
| | | | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | | | - Ann-Louise Esserlind
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anne Francke Christensen
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Folkmann Hansen
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, University of Copenhagen, Roskilde, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Copenhagen, Denmark.,Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen, Copenhagen, Denmark.,iPSYCH-The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | | | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland.,Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Arpo J Aromaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tim Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Marjo-Riitta Järvelin
- Center for Life Course Epidemiology and Systems Medicine, University of Oulu, Oulu, Finland.,Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPE) Centre for Environment and Health, School of Public Health, Imperial College London, London, UK.,Biocenter Oulu, University of Oulu, Oulu, Finland.,Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | | | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David P Strachan
- Population Health Research Institute, St George's, University of London, London, UK
| | - Michel D Ferrari
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Andrea C Belin
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Maija Wessman
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Arn M J M van den Maagdenberg
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands.,Department of Human Genetics, Leiden University Medical Centre, Leiden, the Netherlands
| | - John-Anker Zwart
- FORMI, Oslo University Hospital, Oslo, Norway.,Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | - George Davey Smith
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Kari Stefansson
- deCODE Genetics, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Mark J Daly
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin M Neale
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jes Olesen
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Dale R Nyholt
- Statistical and Genomic Epidemiology Laboratory, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Aarno Palotie
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Arumugam M, Parthasarathy V. Reduction of CD4+CD25+ regulatory T-cells in migraine: Is migraine an autoimmune disorder? J Neuroimmunol 2016; 290:54-9. [DOI: 10.1016/j.jneuroim.2015.11.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 01/08/2023]
|
34
|
Weinstein JJ, Rogers BP, Taylor WD, Boyd BD, Cowan RL, Shelton KM, Salomon RM. Effects of acute tryptophan depletion on raphé functional connectivity in depression. Psychiatry Res 2015; 234:164-71. [PMID: 26411798 PMCID: PMC4631618 DOI: 10.1016/j.pscychresns.2015.08.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 07/21/2015] [Accepted: 08/31/2015] [Indexed: 10/23/2022]
Abstract
Depression remains a great societal burden and a major treatment challenge. Most antidepressant medications target serotonergic raphé nuclei. Acute tryptophan depletion (ATD) modulates serotonin function. To better understand the raphé's role in mood networks, we studied raphé functional connectivity in depression. Fifteen depressed patients were treated with sertraline for 12 weeks and scanned during ATD and sham conditions. Based on our previous findings in a separate cohort, resting state MRI functional connectivity between raphé and other depression-related regions (ROIs) was analyzed in narrow frequency bands. ATD decreased raphé functional connectivity with the bilateral thalamus within 0.025-0.05 Hz, and also decreased raphé functional connectivity with the right pregenual anterior cingulate cortex within 0.05-0.1 Hz. Using the control broadband filter 0.01-0.1 Hz, no significant differences in raphé-ROI functional connectivity were observed. Post-hoc analysis by remission status suggested increased raphé functional connectivity with left pregenual anterior cingulate cortex in remitters (n=10) and decreased raphé functional connectivity with left thalamus in non-remitters (n=5), both within 0.025-0.05 Hz. Reducing serotonin function appears to alter coordination of these mood-related networks in specific, low frequency ranges. For examination of effects of reduced serotonin function on mood-related networks, specific low frequency BOLD fMRI signals can identify regions implicated in neural circuitry and may enable clinically-relevant interpretation of functional connectivity measures. The biological significance of these low frequency signals detected in the raphé merits further study.
Collapse
Affiliation(s)
- Jodi J. Weinstein
- Department of Psychiatry, Vanderbilt University Medical Center
(VUMC), Nashville, TN, USA,Correspondence to: Columbia University Medical Center,
Department of Psychiatry and New York State Psychiatric Institute, 1051 Riverside Drive,
New York, NY 10032, USA
| | - Baxter P. Rogers
- Department of Psychiatry, Vanderbilt University Medical Center
(VUMC), Nashville, TN, USA,Department of Radiology and Radiological Sciences, VUMC,
Nashville, TN, USA,Department of Biomedical Engineering, Vanderbilt University
| | - Warren D. Taylor
- Department of Psychiatry, Vanderbilt University Medical Center
(VUMC), Nashville, TN, USA,The Geriatric Research, Education, and Clinical Center (GRECC),
VA Medical Center, Tennessee Valley Healthcare System, USA
| | - Brian D. Boyd
- Department of Psychiatry, Vanderbilt University Medical Center
(VUMC), Nashville, TN, USA
| | - Ronald L. Cowan
- Department of Psychiatry, Vanderbilt University Medical Center
(VUMC), Nashville, TN, USA,Department of Radiology and Radiological Sciences, VUMC,
Nashville, TN, USA
| | - K. Maureen Shelton
- Department of Psychiatry, Vanderbilt University Medical Center
(VUMC), Nashville, TN, USA
| | - Ronald M. Salomon
- Psychiatric Research Institute, University of Arkansas for
Medical Sciences, Little Rock, AR, USA,Correspondence to: University of Arkansas Medical School
Psychiatric Research Institute, 4301 West Markham Street, Slot 554, Little Rock, AR 72205,
USA. (J.J. Weinstein)
| |
Collapse
|
35
|
Ito K, Kudo M, Sasaki M, Saito A, Yamashita F, Harada T, Yokosawa S, Uwano I, Kameda H, Terayama Y. Detection of changes in the periaqueductal gray matter of patients with episodic migraine using quantitative diffusion kurtosis imaging: preliminary findings. Neuroradiology 2015; 58:115-20. [PMID: 26446146 DOI: 10.1007/s00234-015-1603-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/28/2015] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The periaqueductal gray matter (PAG) is considered to play an important role in generating migraine, but findings from imaging studies remain unclear. Therefore, we investigated whether diffusion kurtosis imaging (DKI) can detect changes in the PAG of migraine patients. METHODS We obtained source images for DKI from 20 patients with episodic migraine and 20 healthy controls using a 3 T magnetic resonance imaging scanner. Mean kurtosis (MK), fractional anisotropy (FA), and mean diffusivity (MD) maps were generated, and the values of the PAG and other deep gray and white matter structures were automatically measured using an atlas-based region-of-interest analysis. The metrics of these structures were compared between the patients and controls. RESULTS The MK and MD values of the PAG were significantly increased in the migraine patients compared with the controls (p < 0.05). The FA values were not significantly different. There were no significant differences in the metrics of the other structures between the patients and controls. The MK values of the PAG were significantly positively correlated with both age and the untreated period in the patient group under univariate analysis (r = 0.53 and 0.56, respectively; p < 0.05) but not multivariate analysis. CONCLUSIONS DKI detected significant increases in the MK and MD values of the PAG in patients with migraine, which suggests that structural changes in the PAG are associated with the pathophysiological mechanisms of migraine.
Collapse
Affiliation(s)
- Kenji Ito
- Division of Ultra-High Field MRI, Institute for Biomedical Sciences, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba, Iwate, 028-3694, Japan.
| | - Masako Kudo
- Department of Neurology and Gerontology, Iwate Medical University, Iwate, Japan
| | - Makoto Sasaki
- Division of Ultra-High Field MRI, Institute for Biomedical Sciences, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba, Iwate, 028-3694, Japan
| | - Ayumi Saito
- Department of Neurology and Gerontology, Iwate Medical University, Iwate, Japan
| | - Fumio Yamashita
- Division of Ultra-High Field MRI, Institute for Biomedical Sciences, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba, Iwate, 028-3694, Japan
| | - Taisuke Harada
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Hokkaido, Japan
| | | | - Ikuko Uwano
- Division of Ultra-High Field MRI, Institute for Biomedical Sciences, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba, Iwate, 028-3694, Japan
| | - Hiroyuki Kameda
- Division of Ultra-High Field MRI, Institute for Biomedical Sciences, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba, Iwate, 028-3694, Japan
| | - Yasuo Terayama
- Department of Neurology and Gerontology, Iwate Medical University, Iwate, Japan
| |
Collapse
|
36
|
Abstract
Cephalic pain and psychiatric disease physiopathology is one of the most elusive issues in medical research, and the cause might be common. Going through the possible reasons of the failure in understanding the physiopathology of these diseases might be helpful to project new studies that might overcome the difficulties encountered and thus open a window on cephalic pain and psychiatric disease. New approaches to psychiatric disease might be applied to cephalic pain.
Collapse
Affiliation(s)
- Alberto E Panerai
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy,
| |
Collapse
|
37
|
The role of purinergic signaling in the etiology of migraine and novel antimigraine treatment. Purinergic Signal 2015; 11:307-16. [PMID: 25957584 PMCID: PMC4529850 DOI: 10.1007/s11302-015-9453-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/28/2015] [Indexed: 12/23/2022] Open
Abstract
Etiopathogenesis of migraine involves different structures of the central nervous system: the trigeminal nerve with nuclei located in the brain stem, vascular system, and the cerebral cortex as well as diverse mechanisms and pathological processes. The multidirectional action of purines in different cell types (blood vessels, neurons, and satellite glial cells) and through different types of purinergic receptors contributes to the etiopathogenesis of migraine pain. Adenosine triphosphate (ATP) and its derivatives are involved in initiation and propagation of migrenogenic signals in several ways: they participate in vasomotor mechanism, cortical spreading depression, and in fast transmission or cross-excitation based on the satellite glial cells in trigeminal ganglion. Contribution of purinergic signaling in the conduction of pain is realized through the activation of P1 and P2 receptors expressed widely in the central nervous system: on the neurons and glial cells as well as on the smooth muscles and endothelium in the vascular system. Therefore, the purinergic receptors can be an excellent target for pharmacologists constructing new antimigraine therapeutics. Moreover, the mechanisms facilitating ATP and adenosine degradation may prevent vasodilatation and thus avoid a secondary central sensitization during a migraine attack. Thus, agonists and antagonists of P receptors as well as ecto-enzymes metabolizing nucleotides/nucleosides could gain the growing attention as therapeutic agents.
Collapse
|
38
|
Gupta SN, Gupta VS, Fields DM. Spectrum of complicated migraine in children: A common profile in aid to clinical diagnosis. World J Clin Pediatr 2015; 4:1-12. [PMID: 25664241 PMCID: PMC4318797 DOI: 10.5409/wjcp.v4.i1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/06/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Complicated migraine encompasses several individual clinical syndromes of migraine. Such a syndrome in children frequently presents with various neurological symptoms in the Emergency Department. An acute presentation in the absence of headache presents a diagnostic challenge. A delay in diagnosis and treatment may have medicolegal implication. To date, there are no reports of a common clinical profile proposed in making a clinical diagnosis for the complicated migraine. In this clinical review, we propose and describe: (1) A common clinical profile in aid to clinical diagnosis for spectrum of complicated migraine; (2) How it can be used in differentiating complicated migraine from migraine without aura, migraine with aura, and seizure; (3) We discuss the status of complicated migraine in the International Headache Society classification 2013; and (4) In addition, a common treatment strategy for the spectrum of migraine has been described. To diagnose complicated migraine clinically, it is imperative to adhere with the proposed profile. This will optimize the use of investigation and will also avoid a legal implication of delay in their management. The proposed common clinical profile is incongruent with the International Headache Society 2013. Future classification should minimize the dissociation from clinically encountered syndromes and coin a single word to address collectively this subtype of migraine with an acute presentation of a common clinical profile.
Collapse
|
39
|
Liu R, Yu S, Li F, Qiu E. Gene expression microarray analysis of the spinal trigeminal nucleus in a rat model of migraine with aura. Neural Regen Res 2015; 7:1931-8. [PMID: 25624821 PMCID: PMC4298885 DOI: 10.3969/j.issn.1673-5374.2012.25.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 08/10/2012] [Indexed: 11/18/2022] Open
Abstract
Cortical spreading depression can trigger migraine with aura and activate the trigeminal vascular system. To examine gene expression profiles in the spinal trigeminal nucleus in rats following cortical spreading depression-induced migraine with aura, a rat model was established by injection of 1 M potassium chloride, which induced cortical spreading depression. DNA microarray analysis revealed that, compared with the control group, the cortical spreading depression group showed seven upregulated genes–myosin heavy chain 1/2, myosin light chain 1, myosin light chain (phosphorylatable, fast skeletal muscle), actin alpha 1, homeobox B8, carbonic anhydrase 3 and an unknown gene. Two genes were downregulated–RGD1563441 and an unknown gene. Real-time quantitative reverse transcription-PCR and bioinformatics analysis indicated that these genes are involved in motility, cell migration, CO2/nitric oxide homeostasis and signal transduction.
Collapse
Affiliation(s)
- Ruozhuo Liu
- Department of Neurology, Chinese PLA General Hospital, Beijing 100853, China
| | - Shengyuan Yu
- Department of Neurology, Chinese PLA General Hospital, Beijing 100853, China
| | - Fengpeng Li
- Department of Neurology, General Hospital of Shenyang Military Region, Shenyang 110016, Liaoning Province, China
| | - Enchao Qiu
- Department of Neurology, the First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100037, China
| |
Collapse
|
40
|
Labruijere S, Chan KY, de Vries R, van den Bogaerdt AJ, Dirven CM, Danser AJ, Kori SH, MaassenVanDenBrink A. Dihydroergotamine and sumatriptan in isolated human coronary artery, middle meningeal artery and saphenous vein. Cephalalgia 2014; 35:182-9. [PMID: 25078720 DOI: 10.1177/0333102414544977] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Dihydroergotamine (DHE) and sumatriptan are contraindicated in patients with cardiovascular disease because of their vasoconstricting properties, which have originally been explored in proximal coronary arteries. Our aim was to investigate DHE and sumatriptan in the proximal and distal coronary artery, middle meningeal artery and saphenous vein. METHODS Blood vessel segments were mounted in organ baths and concentration response curves for DHE and sumatriptan were constructed. RESULTS In the proximal coronary artery, meningeal artery and saphenous vein, maximal contractions to DHE (proximal: 8 ± 4%; meningeal: 32 ± 7%; saphenous: 52 ± 11%) and sumatriptan (proximal: 17 ± 7%; meningeal: 61 ± 18%, saphenous: 37 ± 8%) were not significantly different. In the distal coronary artery, contractions to DHE (5 ± 2%) were significantly smaller than those to sumatriptan (17 ± 9%). At clinically relevant concentrations, mean contractions to DHE and sumatriptan were below 3% in proximal coronary arteries and below 6% in distal coronary arteries. Contractions in the meningeal artery and saphenous vein were higher (7%-38%). CONCLUSIONS Contractions to DHE in distal coronary arteries are smaller than those to sumatriptan, while at clinical concentrations they both induce only slight contractions. In meningeal arteries contractions to DHE and sumatriptan are significantly larger, showing their cranioselectivity. Contractions to DHE in the saphenous vein are higher than those in the arteries, confirming its venous contractile properties.
Collapse
Affiliation(s)
- Sieneke Labruijere
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, The Netherlands
| | - Kayi Y Chan
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, The Netherlands
| | - René de Vries
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, The Netherlands
| | | | - Clemens M Dirven
- Department of Neurosurgery, Erasmus Medical Center, The Netherlands
| | - Ah Jan Danser
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, The Netherlands
| | | | | |
Collapse
|
41
|
Pollak L, Pollak E. Headache during a cluster of benign paroxysmal positional vertigo attacks. Ann Otol Rhinol Laryngol 2014; 123:875-80. [PMID: 25015924 DOI: 10.1177/0003489414539921] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE In view of patients' recurrent complaints, we were interested in investigating the frequency and headache characteristics in patients during a benign paroxysmal positional vertigo (BPPV) cluster. METHODS Patients with BPPV treated at an outpatient dizziness clinic were interviewed about the presence of headache; its quality, localization, severity, time course, and aggravating and alleviating factors; and headache-related disability during their present vertigo cluster. RESULTS Among 152 patients with BPPV, 53 (34.8%) reported headache associated with vertigo. According to The International Classification of Headache Disorders, 8 (15%) patients could be classified as migraine without aura (1.1), 14 (26%) were classified as infrequent episodic tension-type headache associated with pericranial tenderness (2.1.1), 23 (43%) were classified as infrequent episodic tension-type headache without pericranial tenderness (2.1.2), 6 (11%) had cervicogenic headache (11.2.1), and in 2 (4%) patients, the headache could not be specified (14.2). Fifty-two age-matched BPPV patients without headache did not differ in history of headaches, BPPV history, or background diseases. The distribution of canal involvement and number of treatment maneuvers was also similar in both groups. CONCLUSION Headache is frequent in BPPV. The most common is tension-type headache, followed by migraine and cervicogenic headache. Head pain seems to be an independently associated epiphenomenon of BPPV that can worsen patients' distress.
Collapse
Affiliation(s)
- Lea Pollak
- Department of Neurology, Assaf Harofeh Medical Center, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
42
|
[Knowledge of German neurologists on migraine around 1890. Paul Julius Möbius and his 1894 monograph Die Migräne]. DER NERVENARZT 2014; 84:995-1001. [PMID: 23836302 DOI: 10.1007/s00115-013-3826-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Paul Julius Möbius (1853-1907), a Leipzig-based author and editor on a vast majority of subjects, has often been acknowledged as a leading 19th-century German neurologist. His impact on the development of knowledge on migraine has likewise been pointed to. This study compares the monograph published by Möbius on the illness in 1894 with contemporary publications and with present day best practice to establish if the author really made an essential contribution to the problem of migraine. As a representative of the central theory Möbius assumed that migraine was caused by aberrations in the brain. At the same time he made it clear that due to very limited diagnostic options this was only a hypothesis. Apart from a genetic factor and these cerebral changes, for Möbius the general state of health was a decisive factor and prevention and change in lifestyle therefore played a crucial role in his therapeutic recommendations. Basically there were only few differences between the views of Möbius and his colleagues, the major dissimilarity being that Möbius postulated a merely suggestive impact but no physical effect of electrotherapy. Although Möbius's monograph on migraine lacks originality, it provides a concise, easy to understand and stylistically impressive overview on the state of knowledge at that time. Therefore, the book can be considered as a benchmark publication of German speaking neurology around 1890 on migraine and it is highly recommended to present day headache and migraine researchers as well as historians of psychiatry and neurology.
Collapse
|
43
|
Thissen S, Vos IG, Schreuder TH, Schreurs WM, Postma LA, Koehler PJ. Persistent Migraine Aura: New Cases, a Literature Review, and Ideas About Pathophysiology. Headache 2014; 54:1290-309. [DOI: 10.1111/head.12392] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2014] [Indexed: 11/27/2022]
Affiliation(s)
- Sam Thissen
- Department of Neurology; Atrium Medical Centre; Heerlen The Netherlands
| | - Iris G. Vos
- Department of Neurology; Atrium Medical Centre; Heerlen The Netherlands
| | | | - Wendy M.J. Schreurs
- Department of Nuclear Medicine; Atrium Medical Centre; Heerlen The Netherlands
| | - Linda A. Postma
- Department of Neuroradiology; Maastricht University Medical Centre; Maastricht The Netherlands
| | - Peter J. Koehler
- Department of Neurology; Atrium Medical Centre; Heerlen The Netherlands
| |
Collapse
|
44
|
Horváth C. Alterations in brain temperatures as a possible cause of migraine headache. Med Hypotheses 2014; 82:529-34. [PMID: 24581675 DOI: 10.1016/j.mehy.2014.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/24/2014] [Accepted: 02/03/2014] [Indexed: 12/12/2022]
Abstract
Migraine is a debilitating disease with a recurring generally unilateral headache and concomitant symptoms of nausea, vomiting and photo- and/or phonophobia that affects some 11-18% of the population. Most of the mechanisms previously put forward to explain the attacks have been questioned or give an explanation only some of the symptoms. Moreover, the best drugs for treatment are still the 20-year-old triptans, which have serious limitations as regards both efficacy and tolerability. As the dura and some cranial vessels are the only intracranial structures capable of pain sensations, a vascular theory of migraine emerged, but has been debated. Recent theories identified the hyperexcitability of structures involved in pain transmission, such as the trigeminal system or the cortex, or an abnormal modulatory function of the brainstem. However, there is ongoing scientific debate concerning these theories, neither of which is fully capable of explaining the occurrence of a migraine attack. The present article puts forward a hypothesis of the possibility of abnormal temperature regulation in certain regions or the overall brain in migraineurs, the attack being a defense mechanism to prevent neuronal damage. Few examinations have been made of temperature regulation in the human brain. It lacks the carotid rete, a vascular heat exchanger that serves in many animals to provide constant brain temperature. The human brain contains a high density of neurons with a considerable energy demand that is converted to heat. The human brain has a higher temperature than other parts of the body and needs continuous cooling. Recent studies revealed unexpectedly great variations in temperature of various structures of the brain and considerable changes in response to functional activation. There is various evidence in support of the hypothesis that accumulated heat in some structure or the overall brain may be behind the symptoms observed, such as a platelet abnormality, a decreased serotonin content, and dural "inflammation" including vasodilation and brainstem activation. The hypothesis postulates that a migraine attack serves to restore the brain temperature. Abnormally low temperatures in the brain can also result in headache. Surprisingly, no systematic examination of brain temperature changes in migraineurs has been published. Certain case reports support the present hypothesis. Various noninvasive technologies (e.g. MR) capable of monitoring brain temperature are available. If a systematic examination of local brain temperature revealed abnormalities in structures presumed to be involved in migraine, that would increase our understanding of the disease and trigger the development of improved treatment.
Collapse
Affiliation(s)
- Csilla Horváth
- Gedeon Richter Plc., 19-21 Gyömrői út, H-1103 Budapest, Hungary.
| |
Collapse
|
45
|
Greco R, Mangione AS, Siani F, Blandini F, Vairetti M, Nappi G, Sandrini G, Buzzi MG, Tassorelli C. Effects of CGRP receptor antagonism in nitroglycerin-induced hyperalgesia. Cephalalgia 2013; 34:594-604. [DOI: 10.1177/0333102413517776] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 11/28/2013] [Indexed: 11/16/2022]
Abstract
Background The release of calcitonin gene-related peptide (CGRP) from trigeminal nerves plays a central role in the pathophysiology of migraine and clinical evidence shows an antimigraine effect for CGRP receptor antagonists. Systemic administration of nitroglycerin (NTG), a nitrovasodilator, consistently provokes spontaneous-like migraine attacks in migraine sufferers; in the rat, systemic NTG induces a condition of hyperalgesia, probably through the activation of cerebral/spinal structures involved in nociceptive transmission. Aim The aim of this article is to test the analgesic effect of the CGRP receptor antagonist MK-8825 in two animal models of pain that may be relevant for migraine: the tail flick test and the formalin test performed during NTG-induced hyperalgesia. Results MK-8825 showed analgesic activity when administered alone at both the tail flick test and the formalin test. Furthermore, the CGRP antagonist proved effective in counteracting NTG-induced hyperalgesia in both tests. MK-8825 indeed reduced the nociceptive behavior when administered either simultaneously or prior to (30–60 minutes before) NTG. Conclusion These data suggest that MK-8825 may represent a potential therapeutic tool for the treatment of migraine.
Collapse
Affiliation(s)
- R Greco
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
| | - AS Mangione
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
| | - F Siani
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, “C. Mondino” National Neurological Institute, Italy
| | - F Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, “C. Mondino” National Neurological Institute, Italy
| | - M Vairetti
- Department of Internal Medicine and Therapeutics, Pharmacology and Toxicology Unit, University of Pavia, Italy
| | - G Nappi
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
| | - G Sandrini
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
- IRCCS Santa Lucia Foundation, Italy
| | - MG Buzzi
- Department of Brain and Behaviour, University of Pavia, Italy
| | - C Tassorelli
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
- IRCCS Santa Lucia Foundation, Italy
| |
Collapse
|
46
|
Ramírez Rosas MB, Labruijere S, Villalón CM, Maassen Vandenbrink A. Activation of 5-hydroxytryptamine1B/1D/1F receptors as a mechanism of action of antimigraine drugs. Expert Opin Pharmacother 2013; 14:1599-610. [PMID: 23815106 DOI: 10.1517/14656566.2013.806487] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The introduction of the triptans (5-hydroxytryptamine (5-HT)1B/1D receptor agonists) was a great improvement in the acute treatment of migraine. However, shortcomings of the triptans have prompted research on novel serotonergic targets for the treatment of migraine. AREAS COVERED In this review the different types of antimigraine drugs acting at 5-HT receptors, their discovery and development are discussed. The first specific antimigraine drugs were the ergot alkaloids, consisting of ergotamine, dihydroergotamine and methysergide, which are agonists at 5-HT receptors, but can also bind α-adrenoceptors and dopamine receptors. In the 1990s, the triptans became available on the market. They are 5-HT1B/1D receptor agonists, showing fewer side effects due to their receptor specificity. In the last years, compounds that bind specifically to 5-HT1D, 5-HT1F and 5-HT7 receptors have been explored for their antimigraine potential. Furthermore, the serotonergic system seems to act in tight connection with the glutamatergic as well as the CGRP-ergic systems, which may open novel therapeutic avenues. EXPERT OPINION Although the triptans are very effective in treating migraine attacks, their shortcomings have stimulated the search for novel drugs. Currently, the focus is on 5-HT1F receptor agonists, which seem devoid of vascular side effects. Moreover, novel compounds that affect multiple transmitter and/or neuropeptide systems that are involved in migraine could be of therapeutic relevance.
Collapse
Affiliation(s)
- Martha B Ramírez Rosas
- Erasmus Medical Centre, Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
47
|
Abstract
An impaired processing of sensory afferents in the brainstem plays a key role in the development of migraine attack and for many of its clinical aspects. The repetition or prolonged of painful stimuli over time would be able to produce a prolonged and reversible increase of excitability and synaptic efficacy in the nociceptive pathways of the central nervous system. This phenomenon, known as sensitization, involves specifically the caudal trigeminal nucleus. Being an aspect of untreated migraine, allodynia is more common in patients with chronic migraine and migraine with aura, often associated with motor and sensory symptoms sometimes present during the attacks. The presence of allodynia in the course of migraine attack greatly increases the disability of the patient and its recognition, as well as from a therapeutic point of view, it is essential in the management of migraine patients.
Collapse
Affiliation(s)
- Marco Aguggia
- Neurological Department, C. Massaja Hospital, Via Conte Verde 200, 10141 Asti, Italy.
| |
Collapse
|
48
|
Evaluation of c-Fos immunoreactivity in the rat brainstem nuclei relevant in migraine pathogenesis after electrical stimulation of the trigeminal ganglion. Neurol Sci 2013; 34:1597-604. [DOI: 10.1007/s10072-013-1292-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 01/05/2013] [Indexed: 11/25/2022]
|
49
|
Franceschini A, Vilotti S, Ferrari MD, van den Maagdenberg AMJM, Nistri A, Fabbretti E. TNFα levels and macrophages expression reflect an inflammatory potential of trigeminal ganglia in a mouse model of familial hemiplegic migraine. PLoS One 2013; 8:e52394. [PMID: 23326332 PMCID: PMC3543418 DOI: 10.1371/journal.pone.0052394] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 11/13/2012] [Indexed: 01/16/2023] Open
Abstract
Latent changes in trigeminal ganglion structure and function resembling inflammatory conditions may predispose to acute attacks of migraine pain. Here, we investigated whether, in trigeminal sensory ganglia, cytokines such as TNFα might contribute to a local inflammatory phenotype of a transgenic knock-in (KI) mouse model of familial hemiplegic migraine type-1 (FHM-1). To this end, macrophage occurrence and cytokine expression in trigeminal ganglia were compared between wild type (WT) and R192Q mutant CaV2.1 Ca2+ channel (R192Q KI) mice, a genetic model of FHM-1. Cellular and molecular characterization was performed using a combination of confocal immunohistochemistry and cytokine assays. With respect to WT, R192Q KI trigeminal ganglia were enriched in activated macrophages as suggested by their morphology and immunoreactivity to the markers Iba1, CD11b, and ED1. R192Q KI trigeminal ganglia constitutively expressed higher mRNA levels of IL1β, IL6, IL10 and TNFα cytokines and the MCP-1 chemokine. Consistent with the report that TNFα is a major factor to sensitize trigeminal ganglia, we observed that, following an inflammatory reaction evoked by LPS injection, TNFα expression and macrophage occurrence were significantly higher in R192Q KI ganglia with respect to WT ganglia. Our data suggest that, in KI trigeminal ganglia, the complex cellular and molecular environment could support a new tissue phenotype compatible with a neuroinflammatory profile. We propose that, in FHM patients, this condition might contribute to trigeminal pain pathophysiology through release of soluble mediators, including TNFα, that may modulate the crosstalk between sensory neurons and resident glia, underlying the process of neuronal sensitisation.
Collapse
Affiliation(s)
- Alessia Franceschini
- Neuroscience Department, International School for Advanced Studies, Trieste, Italy
| | - Sandra Vilotti
- Neuroscience Department, International School for Advanced Studies, Trieste, Italy
| | - Michel D. Ferrari
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Arn M. J. M. van den Maagdenberg
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Human Genetics, Leiden Genetics University Medical Centre, Leiden, The Netherlands
| | - Andrea Nistri
- Neuroscience Department, International School for Advanced Studies, Trieste, Italy
| | - Elsa Fabbretti
- Neuroscience Department, International School for Advanced Studies, Trieste, Italy
- Center for Biomedical Sciences and Engineering, University of Nova Gorica, Nova Gorica, Slovenia
- * E-mail:
| |
Collapse
|
50
|
Pitarokoili K, Dahlhaus S, Hellwig K, Boehm S, Neubauer H, Gold R, Krogias C. Ventricular tachycardia during basilar-type migraine attack. Ther Adv Neurol Disord 2013; 6:35-40. [PMID: 23277791 DOI: 10.1177/1756285612463625] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Autonomic dysfunction is a characteristic of migraine attacks, rarely, even cardiac repolarization abnormalities have been associated with migraine. We report a case of documented ventricular tachycardia during basilar-type migraine attack. The therapeutic implications of such a co-occurrence as well as a possible relationship between ventricular tachycardia and the underlying biology of basilar-type migraine are discussed.
Collapse
|