1
|
Dalkara T, Kaya Z, Erdener ŞE. Unraveling the interplay of neuroinflammatory signaling between parenchymal and meningeal cells in migraine headache. J Headache Pain 2024; 25:124. [PMID: 39080518 PMCID: PMC11290240 DOI: 10.1186/s10194-024-01827-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The initiation of migraine headaches and the involvement of neuroinflammatory signaling between parenchymal and meningeal cells remain unclear. Experimental evidence suggests that a cascade of inflammatory signaling originating from neurons may extend to the meninges, thereby inducing neurogenic inflammation and headache. This review explores the role of parenchymal inflammatory signaling in migraine headaches, drawing upon recent advancements. BODY: Studies in rodents have demonstrated that sterile meningeal inflammation can stimulate and sensitize meningeal nociceptors, culminating in headaches. The efficacy of relatively blood-brain barrier-impermeable anti-calcitonin gene-related peptide antibodies and triptans in treating migraine attacks, both with and without aura, supports the concept of migraine pain originating in meninges. Additionally, PET studies utilizing inflammation markers have revealed meningeal inflammatory activity in patients experiencing migraine with aura, particularly over the occipital cortex generating visual auras. The parenchymal neuroinflammatory signaling involving neurons, astrocytes, and microglia, which eventually extends to the meninges, can link non-homeostatic perturbations in the insensate brain to pain-sensitive meninges. Recent experimental research has brought deeper insight into parenchymal signaling mechanisms: Neuronal pannexin-1 channels act as stress sensors, initiating the inflammatory signaling by inflammasome formation and high-mobility group box-1 release in response to transient perturbations such as cortical spreading depolarization (CSD) or synaptic metabolic insufficiency caused by transcriptional changes induced by migraine triggers like sleep deprivation and stress. After a single CSD, astrocytes respond by upregulating the transcription of proinflammatory enzymes and mediators, while microglia are involved in restoring neuronal structural integrity; however, repeated CSDs may prompt microglia to adopt a pro-inflammatory state. Transcriptional changes from pro- to anti-inflammatory within 24 h may serve to dampen the inflammatory signaling. The extensive coverage of brain surface and perivascular areas by astrocyte endfeet suggests their role as an interface for transporting inflammatory mediators to the cerebrospinal fluid to contribute to meningeal nociception. CONCLUSION We propose that neuronal stress induced by CSD or synaptic activity-energy mismatch may initiate a parenchymal inflammatory signaling cascade, transmitted to the meninges, thereby triggering lasting headaches characteristic of migraine, with or without aura. This neuroinflammatory interplay between parenchymal and meningeal cells points to the potential for novel targets for migraine treatment and prophylaxis.
Collapse
Affiliation(s)
- Turgay Dalkara
- Departments of Neuroscience and, Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey.
| | - Zeynep Kaya
- Department of Neurology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Şefik Evren Erdener
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
2
|
Kaag Rasmussen M, Møllgård K, Bork PAR, Weikop P, Esmail T, Drici L, Wewer Albrechtsen NJ, Carlsen JF, Huynh NPT, Ghitani N, Mann M, Goldman SA, Mori Y, Chesler AT, Nedergaard M. Trigeminal ganglion neurons are directly activated by influx of CSF solutes in a migraine model. Science 2024; 385:80-86. [PMID: 38963846 DOI: 10.1126/science.adl0544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 05/01/2024] [Indexed: 07/06/2024]
Abstract
Classical migraine patients experience aura, which is transient neurological deficits associated with cortical spreading depression (CSD), preceding headache attacks. It is not currently understood how a pathological event in cortex can affect peripheral sensory neurons. In this study, we show that cerebrospinal fluid (CSF) flows into the trigeminal ganglion, establishing nonsynaptic signaling between brain and trigeminal cells. After CSD, ~11% of the CSF proteome is altered, with up-regulation of proteins that directly activate receptors in the trigeminal ganglion. CSF collected from animals exposed to CSD activates trigeminal neurons in naïve mice in part by CSF-borne calcitonin gene-related peptide (CGRP). We identify a communication pathway between the central and peripheral nervous system that might explain the relationship between migrainous aura and headache.
Collapse
Affiliation(s)
- Martin Kaag Rasmussen
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Peter A R Bork
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tina Esmail
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lylia Drici
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department for Clinical Biochemistry, University Hospital Copenhagen - Bispebjerg, Copenhagen, 2400 Copenhagen, Denmark
| | - Jonathan Frederik Carlsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Radiology, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark
| | - Nguyen P T Huynh
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Sana Biotechnology, Cambridge, MA 02139, USA
| | - Nima Ghitani
- National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD 20892, USA
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Sana Biotechnology, Cambridge, MA 02139, USA
| | - Yuki Mori
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alexander T Chesler
- National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD 20892, USA
- National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen, Denmark
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
3
|
Spekker E, Fejes-Szabó A, Nagy-Grócz G. Models of Trigeminal Activation: Is There an Animal Model of Migraine? Brain Sci 2024; 14:317. [PMID: 38671969 PMCID: PMC11048078 DOI: 10.3390/brainsci14040317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Migraine, recognized as a severe headache disorder, is widely prevalent, significantly impacting the quality of life for those affected. This article aims to provide a comprehensive review of the application of animal model technologies in unraveling the pathomechanism of migraine and developing more effective therapies. It introduces a variety of animal experimental models used in migraine research, emphasizing their versatility and importance in simulating various aspects of the condition. It details the benefits arising from the utilization of these models, emphasizing their role in elucidating pain mechanisms, clarifying trigeminal activation, as well as replicating migraine symptoms and histological changes. In addition, the article consciously acknowledges the inherent limitations and challenges associated with the application of animal experimental models. Recognizing these constraints is a fundamental step toward fine-tuning and optimizing the models for a more accurate reflection of and translatability to the human environment. Overall, a detailed and comprehensive understanding of migraine animal models is crucial for navigating the complexity of the disease. These findings not only provide a deeper insight into the multifaceted nature of migraine but also serve as a foundation for developing effective therapeutic strategies that specifically address the unique challenges arising from migraine pathology.
Collapse
Affiliation(s)
- Eleonóra Spekker
- Interdisciplinary Research Development and Innovation, Center of Excellence, University of Szeged, H-6725 Szeged, Hungary
| | - Annamária Fejes-Szabó
- HUN-REN–SZTE Neuroscience Research Group, University of Szeged, H-6725 Szeged, Hungary;
| | - Gábor Nagy-Grócz
- Department of Theoretical Health Sciences and Health Management, Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári Krt. 31., H-6726 Szeged, Hungary;
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
4
|
Dell’Orco M, Weisend JE, Perrone-Bizzozero NI, Carlson AP, Morton RA, Linsenbardt DN, Shuttleworth CW. Repetitive spreading depolarization induces gene expression changes related to synaptic plasticity and neuroprotective pathways. Front Cell Neurosci 2023; 17:1292661. [PMID: 38162001 PMCID: PMC10757627 DOI: 10.3389/fncel.2023.1292661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024] Open
Abstract
Spreading depolarization (SD) is a slowly propagating wave of profound depolarization that sweeps through cortical tissue. While much emphasis has been placed on the damaging consequences of SD, there is uncertainty surrounding the potential activation of beneficial pathways such as cell survival and plasticity. The present study used unbiased assessments of gene expression to evaluate that compensatory and repair mechanisms could be recruited following SD, regardless of the induction method, which prior to this work had not been assessed. We also tested assumptions of appropriate controls and the spatial extent of expression changes that are important for in vivo SD models. SD clusters were induced with either KCl focal application or optogenetic stimulation in healthy mice. Cortical RNA was extracted and sequenced to identify differentially expressed genes (DEGs). SDs using both induction methods significantly upregulated 16 genes (vs. sham animals) that included the cell proliferation-related genes FOS, JUN, and DUSP6, the plasticity-related genes ARC and HOMER1, and the inflammation-related genes PTGS2, EGR2, and NR4A1. The contralateral hemisphere is commonly used as control tissue for DEG studies, but its activity could be modified by near-global disruption of activity in the adjacent brain. We found 21 upregulated genes when comparing SD-involved cortex vs. tissue from the contralateral hemisphere of the same animals. Interestingly, there was almost complete overlap (21/16) with the DEGs identified using sham controls. Neuronal activity also differs in SD initiation zones, where sustained global depolarization is required to initiate propagating events. We found that gene expression varied as a function of the distance from the SD initiation site, with greater expression differences observed in regions further away. Functional and pathway enrichment analyses identified axonogenesis, branching, neuritogenesis, and dendritic growth as significantly enriched in overlapping DEGs. Increased expression of SD-induced genes was also associated with predicted inhibition of pathways associated with cell death, and apoptosis. These results identify novel biological pathways that could be involved in plasticity and/or circuit modification in brain tissue impacted by SD. These results also identify novel functional targets that could be tested to determine potential roles in the recovery and survival of peri-infarct tissues.
Collapse
Affiliation(s)
- Michela Dell’Orco
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jordan E. Weisend
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Nora I. Perrone-Bizzozero
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Andrew P. Carlson
- Department of Neurosurgery, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Russell A. Morton
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - David N. Linsenbardt
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - C. William Shuttleworth
- Department of Neurosciences, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
5
|
Ebrahimi A, Kamyab A, Hosseini S, Ebrahimi S, Ashkani-Esfahani S. Involvement of Coenzyme Q10 in Various Neurodegenerative and Psychiatric Diseases. Biochem Res Int 2023; 2023:5510874. [PMID: 37946741 PMCID: PMC10632062 DOI: 10.1155/2023/5510874] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/06/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Coenzyme Q10 (CoQ10), commonly known as ubiquinone, is a vitamin-like component generated in mitochondrial inner membranes. This molecule is detected broadly in different parts of the human body in various quantities. This molecule can be absorbed by the digestive system from various nutritional sources as supplements. CoQ10 exists in three states: in a of reduced form (ubiquinol), in a semiquinone radical form, and in oxidized ubiquinone form in different organs of the body, playing a crucial role in electron transportation and contributing to energy metabolism and oxygen utilization, especially in the musculoskeletal and nervous systems. Since the early 1980s, research about CoQ10 has become the interest for two reasons. First, CoQ10 deficiency has been found to have a link with cardiovascular, neurologic, and cancer disorders. Second, this molecule has an antioxidant and free-radical scavenger nature. Since then, several investigations have indicated that the drug may benefit patients with cardiovascular, neuromuscular, and neurodegenerative illnesses. CoQ10 may protect the neurological system from degeneration and degradation due to its antioxidant and energy-regulating activity in mitochondria. This agent has shown its efficacy in preventing and treating neurological diseases such as migraine, Parkinson's disease, Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, and Friedreich's ataxia. This study reviews the literature to highlight this agent's potential therapeutic effects in the mentioned neurological disorders.
Collapse
Affiliation(s)
- Alireza Ebrahimi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Sahar Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Ebrahimi
- Department of Medical Ethics, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
6
|
Rubin M, Cutillo G, Zanandrea L, Montini F, Zanetta C, Bellini A, Cecchetti G, Fanelli GF, Falini A, Scotti R, Calloni SF, Di Bella D, Filippi M, Colombo B. Crossing the border between epileptic and vascular pathology: a report of CACNA1A-related treatment-resistant hemiplegic migraine. J Neurol 2023; 270:5639-5644. [PMID: 37466662 DOI: 10.1007/s00415-023-11877-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/20/2023]
Affiliation(s)
- M Rubin
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - G Cutillo
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - L Zanandrea
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - F Montini
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - C Zanetta
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - A Bellini
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - G Cecchetti
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - G F Fanelli
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - A Falini
- Department of Neuroradiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - R Scotti
- Department of Neuroradiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S F Calloni
- Department of Neuroradiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - D Di Bella
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - M Filippi
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy.
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - B Colombo
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132, Milan, Italy
| |
Collapse
|
7
|
Dell’Orco M, Weisend JE, Perrone-Bizzozero NI, Carlson AP, Morton RA, Linsenbardt DN, Shuttleworth CW. Repetitive Spreading Depolarization induces gene expression changes related to synaptic plasticity and neuroprotective pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530317. [PMID: 36909568 PMCID: PMC10002705 DOI: 10.1101/2023.02.27.530317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Spreading depolarization (SD) is a slowly propagating wave of profound depolarization that sweeps through cortical tissue. While much emphasis has been placed on the damaging consequences of SD, there is uncertainty surrounding the potential activation of beneficial pathways such as cell survival and plasticity. The present study used unbiased assessments of gene expression to evaluate that compensatory and repair mechanisms could be recruited following SD, regardless of the induction method, which prior to this work had not been assessed. We also tested assumptions of appropriate controls and the spatial extent of expression changes that are important for in vivo SD models. SD clusters were induced with either KCl focal application or optogenetic stimulation in healthy mice. Cortical RNA was extracted and sequenced to identify differentially expressed genes (DEGs). SDs using both induction methods significantly upregulated 16 genes (versus sham animals) that included the cell proliferation-related genes FOS, JUN, and DUSP6, the plasticity-related genes ARC and HOMER1, and the inflammation-related genes PTGS2, EGR2, and NR4A1. The contralateral hemisphere is commonly used as control tissue for DEG studies, but its activity could be modified by near-global disruption of activity in the adjacent brain. We found 21 upregulated genes when comparing SD-involved cortex versus tissue from the contralateral hemisphere of the same animals. Interestingly, there was almost complete overlap (21/16) with the DEGs identified using sham controls. Neuronal activity also differs in SD initiation zones, where sustained global depolarization is required to initiate propagating events. We found that gene expression varied as a function of the distance from the SD initiation site, with greater expression differences observed in regions further away. Functional and pathway enrichment analyses identified axonogenesis, branching, neuritogenesis, and dendritic growth as significantly enriched in overlapping DEGs. Increased expression of SD-induced genes was also associated with predicted inhibition of pathways associated with cell death, and apoptosis. These results identify novel biological pathways that could be involved in plasticity and/or circuit modification in brain tissue impacted by SD. These results also identify novel functional targets that could be tested to determine potential roles in recovery and survival of peri-infarct tissues.
Collapse
Affiliation(s)
- Michela Dell’Orco
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - Jordan E. Weisend
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - Nora I. Perrone-Bizzozero
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - Andrew P. Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - Russell A. Morton
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - David N Linsenbardt
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, 87131, USA
| |
Collapse
|
8
|
Dehghani A, Schenke M, van Heiningen SH, Karatas H, Tolner EA, van den Maagdenberg AMJM. Optogenetic cortical spreading depolarization induces headache-related behaviour and neuroinflammatory responses some prolonged in familial hemiplegic migraine type 1 mice. J Headache Pain 2023; 24:96. [PMID: 37495957 PMCID: PMC10373261 DOI: 10.1186/s10194-023-01628-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Cortical spreading depolarization (CSD), the neurophysiological correlate of the migraine aura, can activate trigeminal pain pathways, but the neurobiological mechanisms and behavioural consequences remain unclear. Here we investigated effects of optogenetically-induced CSDs on headache-related behaviour and neuroinflammatory responses in transgenic mice carrying a familial hemiplegic migraine type 1 (FHM1) mutation. METHODS CSD events (3 in total) were evoked in a minimally invasive manner by optogenetic stimulation through the intact skull in freely behaving wildtype (WT) and FHM1 mutant mice. Related behaviours were analysed using mouse grimace scale (MGS) scoring, head grooming, and nest building behaviour. Neuroinflammatory changes were investigated by assessing HMGB1 release with immunohistochemistry and by pre-treating mice with a selective Pannexin-1 channel inhibitor. RESULTS In both WT and FHM1 mutant mice, CSDs induced headache-related behaviour, as evidenced by increased MGS scores and the occurrence of oculotemporal strokes, at 30 min. Mice of both genotypes also showed decreased nest building behaviour after CSD. Whereas in WT mice MGS scores had normalized at 24 h after CSD, in FHM1 mutant mice scores were normalized only at 48 h. Of note, oculotemporal stroke behaviour already normalized 5 h after CSD, whereas nest building behaviour remained impaired at 72 h; no genotype differences were observed for either readout. Nuclear HMGB1 release in the cortex of FHM1 mutant mice, at 30 min after CSD, was increased bilaterally in both WT and FHM1 mutant mice, albeit that contralateral release was more pronounced in the mutant mice. Only in FHM1 mutant mice, contralateral release remained higher at 24 h after CSD, but at 48 h had returned to abnormal, elevated, baseline values, when compared to WT mice. Blocking Panx1 channels by TAT-Panx308 inhibited CSD-induced headache related behaviour and HMGB1 release. CONCLUSIONS CSDs, induced in a minimally invasive manner by optogenetics, investigated in freely behaving mice, cause various migraine relevant behavioural and neuroinflammatory phenotypes that are more pronounced and longer-lasting in FHM1 mutant compared to WT mice. Prevention of CSD-related neuroinflammatory changes may have therapeutic potential in the treatment of migraine.
Collapse
Affiliation(s)
- Anisa Dehghani
- Department of Human Genetics, Leiden University Medical Center, Leiden, RC, 2300, The Netherlands.
- Department of Anesthesia and Critical Care and Pain Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Maarten Schenke
- Department of Human Genetics, Leiden University Medical Center, Leiden, RC, 2300, The Netherlands
| | - Sandra H van Heiningen
- Department of Human Genetics, Leiden University Medical Center, Leiden, RC, 2300, The Netherlands
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Else A Tolner
- Department of Human Genetics, Leiden University Medical Center, Leiden, RC, 2300, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, RC, 2300, The Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, RC, 2300, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, RC, 2300, The Netherlands.
| |
Collapse
|
9
|
Mehra A, Gomez F, Bischof H, Diedrich D, Laudanski K. Cortical Spreading Depolarization and Delayed Cerebral Ischemia; Rethinking Secondary Neurological Injury in Subarachnoid Hemorrhage. Int J Mol Sci 2023; 24:9883. [PMID: 37373029 DOI: 10.3390/ijms24129883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Poor outcomes in Subarachnoid Hemorrhage (SAH) are in part due to a unique form of secondary neurological injury known as Delayed Cerebral Ischemia (DCI). DCI is characterized by new neurological insults that continue to occur beyond 72 h after the onset of the hemorrhage. Historically, it was thought to be a consequence of hypoperfusion in the setting of vasospasm. However, DCI was found to occur even in the absence of radiographic evidence of vasospasm. More recent evidence indicates that catastrophic ionic disruptions known as Cortical Spreading Depolarizations (CSD) may be the culprits of DCI. CSDs occur in otherwise healthy brain tissue even without demonstrable vasospasm. Furthermore, CSDs often trigger a complex interplay of neuroinflammation, microthrombi formation, and vasoconstriction. CSDs may therefore represent measurable and modifiable prognostic factors in the prevention and treatment of DCI. Although Ketamine and Nimodipine have shown promise in the treatment and prevention of CSDs in SAH, further research is needed to determine the therapeutic potential of these as well as other agents.
Collapse
Affiliation(s)
- Ashir Mehra
- Department of Neurology, University of Missouri, Columbia, MO 65212, USA
| | - Francisco Gomez
- Department of Neurology, University of Missouri, Columbia, MO 65212, USA
| | - Holly Bischof
- Penn Presbyterian Medical Center, Philadelphia, PA 19104, USA
| | - Daniel Diedrich
- Department of Anesthesiology and Perioperative Care, Mayo Clinic, Rochester, MN 55905, USA
| | - Krzysztof Laudanski
- Department of Anesthesiology and Perioperative Care, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
10
|
Yamanaka G, Hayashi K, Morishita N, Takeshita M, Ishii C, Suzuki S, Ishimine R, Kasuga A, Nakazawa H, Takamatsu T, Watanabe Y, Morichi S, Ishida Y, Yamazaki T, Go S. Experimental and Clinical Investigation of Cytokines in Migraine: A Narrative Review. Int J Mol Sci 2023; 24:ijms24098343. [PMID: 37176049 PMCID: PMC10178908 DOI: 10.3390/ijms24098343] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
The role of neuroinflammation in the pathophysiology of migraines is increasingly being recognized, and cytokines, which are important endogenous substances involved in immune and inflammatory responses, have also received attention. This review examines the current literature on neuroinflammation in the pathogenesis of migraine. Elevated TNF-α, IL-1β, and IL-6 levels have been identified in non-invasive mouse models with cortical spreading depolarization (CSD). Various mouse models to induce migraine attack-like symptoms also demonstrated elevated inflammatory cytokines and findings suggesting differences between episodic and chronic migraines and between males and females. While studies on human blood during migraine attacks have reported no change in TNF-α levels and often inconsistent results for IL-1β and IL-6 levels, serial analysis of cytokines in jugular venous blood during migraine attacks revealed consistently increased IL-1β, IL-6, and TNF-α. In a study on the interictal period, researchers reported higher levels of TNF-α and IL-6 compared to controls and no change regarding IL-1β levels. Saliva-based tests suggest that IL-1β might be useful in discriminating against migraine. Patients with migraine may benefit from a cytokine perspective on the pathogenesis of migraine, as there have been several encouraging reports suggesting new therapeutic avenues.
Collapse
Affiliation(s)
- Gaku Yamanaka
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Kanako Hayashi
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Natsumi Morishita
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Mika Takeshita
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Chiako Ishii
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Shinji Suzuki
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Rie Ishimine
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Akiko Kasuga
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Haruka Nakazawa
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Tomoko Takamatsu
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Yusuke Watanabe
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Shinichiro Morichi
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Yu Ishida
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Takashi Yamazaki
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Soken Go
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| |
Collapse
|
11
|
Biscetti L, Cresta E, Cupini LM, Calabresi P, Sarchielli P. The putative role of neuroinflammation in the complex pathophysiology of migraine: From bench to bedside. Neurobiol Dis 2023; 180:106072. [PMID: 36907522 DOI: 10.1016/j.nbd.2023.106072] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/18/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
The implications of neurogenic inflammation and neuroinflammation in the pathophysiology of migraine have been clearly demonstrated in preclinical migraine models involving several sites relevant in the trigemino-vascular system, including dural vessels and trigeminal endings, the trigeminal ganglion, the trigeminal nucleus caudalis as well as central trigeminal pain processing structures. In this context, a relevant role has been attributed over the years to some sensory and parasympathetic neuropeptides, in particular calcitonin gene neuropeptide, vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide. Several preclinical and clinical lines of evidence also support the implication of the potent vasodilator and messenger molecule nitric oxide in migraine pathophysiology. All these molecules are involved in vasodilation of the intracranial vasculature, as well as in the peripheral and central sensitization of the trigeminal system. At meningeal level, the engagement of some immune cells of innate immunity, including mast-cells and dendritic cells, and their mediators, has been observed in preclinical migraine models of neurogenic inflammation in response to sensory neuropeptides release due to trigemino-vascular system activation. In the context of neuroinflammatory events implicated in migraine pathogenesis, also activated glial cells in the peripheral and central structures processing trigeminal nociceptive signals seem to play a relevant role. Finally, cortical spreading depression, the pathophysiological substrate of migraine aura, has been reported to be associated with inflammatory mechanisms such as pro-inflammatory cytokine upregulation and intracellular signalling. Reactive astrocytosis consequent to cortical spreading depression is linked to an upregulation of these inflammatory markers. The present review summarizes current findings on the roles of immune cells and inflammatory responses in the pathophysiology of migraine and their possible exploitation in the view of innovative disease-modifying strategies.
Collapse
Affiliation(s)
- Leonardo Biscetti
- Istituto Nazionale di Ricovero e Cura dell'Anziano a carattere scientifico, IRCCS-INRCA, Ancona, Italy.
| | - Elena Cresta
- 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
|
12
|
Salahi M, Parsa S, Nourmohammadi D, Razmkhah Z, Salimi O, Rahmani M, Zivary S, Askarzadeh M, Tapak MA, Vaezi A, Sadeghsalehi H, Yaghoobpoor S, Mottahedi M, Garousi S, Deravi N. Immunologic aspects of migraine: A review of literature. Front Neurol 2022; 13:944791. [PMID: 36247795 PMCID: PMC9554313 DOI: 10.3389/fneur.2022.944791] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022] Open
Abstract
Migraine headaches are highly prevalent, affecting 15% of the population. However, despite many studies to determine this disease's mechanism and efficient management, its pathophysiology has not been fully elucidated. There are suggested hypotheses about the possible mediating role of mast cells, immunoglobulin E, histamine, and cytokines in this disease. A higher incidence of this disease in allergic and asthma patients, reported by several studies, indicates the possible role of brain mast cells located around the brain vessels in this disease. The mast cells are more specifically within the dura and can affect the trigeminal nerve and cervical or sphenopalatine ganglion, triggering the secretion of substances that cause migraine. Neuropeptides such as calcitonin gene-related peptide (CGRP), neurokinin-A, neurotensin (NT), pituitary adenylate-cyclase-activating peptide (PACAP), and substance P (SP) trigger mast cells, and in response, they secrete pro-inflammatory and vasodilatory molecules such as interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) as a selective result of corticotropin-releasing hormone (CRH) secretion. This stress hormone contributes to migraine or intensifies it. Blocking these pathways using immunologic agents such as CGRP antibody, anti-CGRP receptor antibody, and interleukin-1 beta (IL-1β)/interleukin 1 receptor type 1 (IL-1R1) axis-related agents may be promising as potential prophylactic migraine treatments. This review is going to summarize the immunological aspects of migraine.
Collapse
Affiliation(s)
- Mehrnaz Salahi
- Student Research Committee, School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sina Parsa
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Delaram Nourmohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Razmkhah
- Student Research Committee, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Salimi
- Student Research Committee, Faculty of Medicine, Islamic Azad University of Najafabad, Isfahan, Iran
| | | | - Saeid Zivary
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Monireh Askarzadeh
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Tapak
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ali Vaezi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Sadeghsalehi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shirin Yaghoobpoor
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehran Mottahedi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Setareh Garousi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Changes in Plasma Lipid Levels Following Cortical Spreading Depolarization in a Transgenic Mouse Model of Familial Hemiplegic Migraine. Metabolites 2022; 12:metabo12030220. [PMID: 35323663 PMCID: PMC8953552 DOI: 10.3390/metabo12030220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 01/01/2023] Open
Abstract
Metabolite levels in peripheral body fluids can correlate with attack features in migraine patients, which underscores the potential of plasma metabolites as possible disease biomarkers. Migraine headache can be preceded by an aura that is caused by cortical spreading depolarization (CSD), a transient wave of neuroglial depolarization. We previously identified plasma amino acid changes after CSD in familial hemiplegic migraine type 1 (FHM1) mutant mice that exhibit increased neuronal excitability and various migraine-related features. Here, we aimed to uncover lipid metabolic pathways affected by CSD, guided by findings on the involvement of lipids in hemiplegic migraine pathophysiology. Using targeted lipidomic analysis, we studied plasma lipid metabolite levels at different time points after CSD in wild-type and FHM1 mutant mice. Following CSD, the most prominent plasma lipid change concerned a transient increase in PGD2, which lasted longer in mutant mice. In wild-type mice only, levels of anti-inflammatory lipid mediators DPAn-3, EPA, ALA, and DHA were elevated 24 h following CSD compared to Sham-treated animals. Given the role of PGs and neuroinflammation in migraine pathophysiology, our findings underscore the potential of monitoring peripheral changes in lipids to gain insight in central brain mechanisms.
Collapse
|
14
|
Eren-Koçak E, Dalkara T. Ion Channel Dysfunction and Neuroinflammation in Migraine and Depression. Front Pharmacol 2021; 12:777607. [PMID: 34858192 PMCID: PMC8631474 DOI: 10.3389/fphar.2021.777607] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 01/15/2023] Open
Abstract
Migraine and major depression are debilitating disorders with high lifetime prevalence rates. Interestingly these disorders are highly comorbid and show significant heritability, suggesting shared pathophysiological mechanisms. Non-homeostatic function of ion channels and neuroinflammation may be common mechanisms underlying both disorders: The excitation-inhibition balance of microcircuits and their modulation by monoaminergic systems, which depend on the expression and function of membrane located K+, Na+, and Ca+2 channels, have been reported to be disturbed in both depression and migraine. Ion channels and energy supply to synapses not only change excitability of neurons but can also mediate the induction and maintenance of inflammatory signaling implicated in the pathophysiology of both disorders. In this respect, Pannexin-1 and P2X7 large-pore ion channel receptors can induce inflammasome formation that triggers release of pro-inflammatory mediators from the cell. Here, the role of ion channels involved in the regulation of excitation-inhibition balance, synaptic energy homeostasis as well as inflammatory signaling in migraine and depression will be reviewed.
Collapse
Affiliation(s)
- Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Psychiatry, Medical Faculty, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
15
|
Chen Y, Wang S, Wang Y. Role of herbal medicine for prevention and treatment of migraine. Phytother Res 2021; 36:730-760. [PMID: 34818682 DOI: 10.1002/ptr.7339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/06/2021] [Accepted: 11/02/2021] [Indexed: 01/18/2023]
Abstract
Migraine is a disabling neurovascular disease with unilateral or bilateral pulsatile headache, which intensively affects human health and quality of life due to high morbidity worldwide. Migraine is commonly accompanied by abnormal pain sensitization, neuroinflammatory response, and vasomotor dysfunction. Owing to the management dilemmas of migraine, there is an urgent need to develop effective and low-cost therapies. In recent years, herbal medicines as a promising strategy with analgesic activity and minor side effect, have been proposed for the prevention and treatment of migraine. Considering the lack of a review integrating experimental studies regarding the herbal treatment of migraine, this review systematically summarizes the important potential applications of herbal medicines in ameliorating migraine via multiple therapeutic targets and pathways, as well as provides a reference for further development of novel antimigraine drugs.
Collapse
Affiliation(s)
- Yulong Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.,Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
16
|
Erdener ŞE, Kaya Z, Dalkara T. Parenchymal neuroinflammatory signaling and dural neurogenic inflammation in migraine. J Headache Pain 2021; 22:138. [PMID: 34794382 PMCID: PMC8600694 DOI: 10.1186/s10194-021-01353-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background Pain is generally concomitant with an inflammatory reaction at the site where the nociceptive fibers are activated. Rodent studies suggest that a sterile meningeal inflammatory signaling cascade may play a role in migraine headache as well. Experimental studies also suggest that a parenchymal inflammatory signaling cascade may report the non-homeostatic conditions in brain to the meninges to induce headache. However, how these signaling mechanisms function in patients is unclear and debated. Our aim is to discuss the role of inflammatory signaling in migraine pathophysiology in light of recent developments. Body Rodent studies suggest that a sterile meningeal inflammatory reaction can be initiated by release of peptides from active trigeminocervical C-fibers and stimulation of resident macrophages and dendritic/mast cells. This inflammatory reaction might be needed for sustained stimulation and sensitization of meningeal nociceptors after initial activation along with ganglionic and central mechanisms. Most migraines likely have cerebral origin as suggested by prodromal neurologic symptoms. Based on rodent studies, a parenchymal inflammatory signaling cascade has been proposed as a potential mechanism linking cortical spreading depolarization (CSD) to meningeal nociception. A recent PET/MRI study using a sensitive inflammation marker showed the presence of meningeal inflammatory activity in migraine with aura patients over the occipital cortex generating the visual aura. These studies also suggest the presence of a parenchymal inflammatory activity, supporting the experimental findings. In rodents, parenchymal inflammatory signaling has also been shown to be activated by migraine triggers such as sleep deprivation without requiring a CSD because of the resultant transcriptional changes, predisposing to inadequate synaptic energy supply during intense excitatory transmission. Thus, it may be hypothesized that neuronal stress created by either CSD or synaptic activity-energy mismatch could both initiate a parenchymal inflammatory signaling cascade, propagating to the meninges, where it is converted to a lasting headache with or without aura. Conclusion Experimental studies in animals and emerging imaging findings from patients warrant further research to gain deeper insight to the complex role of inflammatory signaling in headache generation in migraine.
Collapse
Affiliation(s)
- Şefik Evren Erdener
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Zeynep Kaya
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sıhhiye, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, and Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
17
|
Bauer PR, Tolner EA, Keezer MR, Ferrari MD, Sander JW. Headache in people with epilepsy. Nat Rev Neurol 2021; 17:529-544. [PMID: 34312533 DOI: 10.1038/s41582-021-00516-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Epidemiological estimates indicate that individuals with epilepsy are more likely to experience headaches, including migraine, than individuals without epilepsy. Headaches can be temporally unrelated to seizures, or can occur before, during or after an episode; seizures and migraine attacks are mostly not temporally linked. The pathophysiological links between headaches (including migraine) and epilepsy are complex and have not yet been fully elucidated. Correct diagnoses and appropriate treatment of headaches in individuals with epilepsy is essential, as headaches can contribute substantially to disease burden. Here, we review the insights that have been made into the associations between headache and epilepsy over the past 5 years, including information on the pathophysiological mechanisms and genetic variants that link the two disorders. We also discuss the current best practice for the management of headaches co-occurring with epilepsy and highlight future challenges for this area of research.
Collapse
Affiliation(s)
- Prisca R Bauer
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Freiburg, Freiburg, Germany.
| | - Else A Tolner
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Mark R Keezer
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,School of Public Health, Université de Montréal, Montreal, Quebec, Canada.,Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands
| | - Michel D Ferrari
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Josemir W Sander
- Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands.,NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London, UK.,Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| |
Collapse
|
18
|
Freitag FG. Headache Medicine Grand Challenge: Headache: A New Frontier, A New Challenge. FRONTIERS IN PAIN RESEARCH 2021; 2:690683. [PMID: 35295521 PMCID: PMC8915632 DOI: 10.3389/fpain.2021.690683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/04/2021] [Indexed: 11/26/2022] Open
Affiliation(s)
- Frederick G. Freitag
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
19
|
Dehghani A, Phisonkunkasem T, Yilmaz Ozcan S, Dalkara T, van den Maagdenberg AMJM, Tolner EA, Karatas H. Widespread brain parenchymal HMGB1 and NF-κB neuroinflammatory responses upon cortical spreading depolarization in familial hemiplegic migraine type 1 mice. Neurobiol Dis 2021; 156:105424. [PMID: 34118418 DOI: 10.1016/j.nbd.2021.105424] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/04/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammatory changes involving neuronal HMGB1 release and astrocytic NF-κB nuclear translocation occur following cortical spreading depolarization (CSD) in wildtype (WT) mice but it is unknown to what extent this occurs in the migraine brain. We therefore investigated in familial hemiplegic migraine type 1 (FHM1) knock-in mice, which express an intrinsic hyperexcitability phenotype, the extent of neuroinflammation without and after CSD. CSD was evoked in one hemisphere by pinprick (single CSD) or topical KCl application (multiple CSDs). Neuroinflammatory (HMGB1, NF-κB) and neuronal activation (pERK) markers were investigated by immunohistochemistry in the brains of WT and FHM1 mutant mice without and after CSD. Effects of NMDA receptor antagonism on basal and CSD-induced neuroinflammatory changes were examined by, respectively, systemically administered MK801 and ifenprodil or topical MK801 application. In FHM1 mutant mice, CSD caused enhanced neuronal HMGB1 release and astrocytic NF-κB nuclear translocation in the cortex and subcortical areas that were equally high in both hemispheres. In WT mice such effects were only pronounced in the hemisphere in which CSD was induced. Neuroinflammatory responses were associated with pERK expression indicating neuronal activation. Upon CSD, contralateral cortical and striatal HMGB1 release was reduced by topical application of MK801 in the hemisphere contralateral to the one in which CSD was induced. This study reveals that neuroinflammatory activation after CSD is widespread and extends to the contralateral hemisphere, particularly in brains of FHM1 mutant mice. Effective blockade of CSD-induced neuroinflammatory responses in the contralateral hemisphere in FHM1 mice by local NMDA receptor antagonism suggests that neuronal hyperexcitability-related neuroinflammation is relevant in migraine pathophysiology, but possibly also other neurological disorders in which spreading depolarization is involved.
Collapse
Affiliation(s)
- Anisa Dehghani
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey; Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.
| | - Thas Phisonkunkasem
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sinem Yilmaz Ozcan
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Else A Tolner
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
20
|
Kursun O, Yemisci M, van den Maagdenberg AMJM, Karatas H. Migraine and neuroinflammation: the inflammasome perspective. J Headache Pain 2021; 22:55. [PMID: 34112082 PMCID: PMC8192049 DOI: 10.1186/s10194-021-01271-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neuroinflammation has an important role in the pathophysiology of migraine, which is a complex neuro-glio-vascular disorder. The main aim of this review is to highlight findings of cortical spreading depolarization (CSD)-induced neuroinflammatory signaling in brain parenchyma from the inflammasome perspective. In addition, we discuss the limited data of the contribution of inflammasomes to other aspects of migraine pathophysiology, foremost the activation of the trigeminovascular system and thereby the generation of migraine pain. MAIN BODY Inflammasomes are signaling multiprotein complexes and key components of the innate immune system. Their activation causes the production of inflammatory cytokines that can stimulate trigeminal neurons and are thus relevant to the generation of migraine pain. The contribution of inflammasome activation to pain signaling has attracted considerable attention in recent years. Nucleotide-binding domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) is the best characterized inflammasome and there is emerging evidence of its role in a variety of inflammatory pain conditions, including migraine. In this review, we discuss, from an inflammasome point of view, cortical spreading depolarization (CSD)-induced neuroinflammatory signaling in brain parenchyma, the connection with genetic factors that make the brain vulnerable to CSD, and the relation of the inflammasome with diseases that are co-morbid with migraine, including stroke, epilepsy, and the possible links with COVID-19 infection. CONCLUSION Neuroinflammatory pathways, specifically those involving inflammasome proteins, seem promising candidates as treatment targets, and perhaps even biomarkers, in migraine.
Collapse
Affiliation(s)
| | - Muge Yemisci
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands.,Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
21
|
Sueiras M, Thonon V, Santamarina E, Sánchez-Guerrero Á, Poca MA, Quintana M, Riveiro M, Sahuquillo J. Cortical Spreading Depression Phenomena Are Frequent in Ischemic and Traumatic Penumbra: A Prospective Study in Patients With Traumatic Brain Injury and Large Hemispheric Ischemic Stroke. J Clin Neurophysiol 2021; 38:47-55. [PMID: 31702708 DOI: 10.1097/wnp.0000000000000648] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
PURPOSE Spreading depolarization (SD) phenomena are waves of neuronal depolarization, which propagate slowly at a velocity of 1 to 5 mm/minute and can occur in patients with ischemic or hemorrhagic stroke, traumatic brain injury, and migraine with aura. They form part of secondary injury, occurring after spreading ischemia. The purposes of this study were to describe the frequency and characteristics of SD phenomena and to define whether a correlation existed between SD and outcome in a group of patients with TBI and large hemispheric ischemic stroke. METHODS This was a prospective observational study of 39 adult patients, 17 with malignant middle cerebral artery infarction and 22 with moderate or severe traumatic brain injury, who underwent decompressive craniectomy and multimodal neuromonitoring including electrocorticography. Identification, classification, and interpretation of SDs were performed using the published recommendations from the Cooperative Study on Brain Injury Depolarization group. The outcomes assessed were functional disability at 6 and 12 months after injury, according to the extended Glasgow outcome scale, Barthel index, and modified Rankin scale. RESULTS Four hundred eighty-three SDs were detected, in 58.9% of the patients. Spreading depolarizations were more common, particularly the isoelectric SD type, in patients with malignant middle cerebral artery infarction (P < 0.04). In 65.21% of patients with SDs on electrocorticography, the "peak" day of depolarization was day 0 (the first 24 hours of recording). Spreading depolarization convulsions were present in 26.08% of patients with SDs. Patients with more SDs and higher depolarization indices scored worse on extended Glasgow outcome scale (6 months) and Barthel index (6 and 12 months) (P < 0.05). CONCLUSIONS Evidence on SD phenomena is important to ensure continued progress in understanding their pathophysiology, in the search for therapeutic targets to avoid additional damage from these secondary injuries.
Collapse
Affiliation(s)
- Maria Sueiras
- Department of Clinical Neurophysiology, Vall d'Hebron University Hospital, Barcelona, Spain
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Vanessa Thonon
- Department of Clinical Neurophysiology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Estevo Santamarina
- Epilepsy Unit, Department of Neurology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Ángela Sánchez-Guerrero
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Maria A Poca
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Department of Neurosurgery, Vall d'Hebron University Hospital, Barcelona, Spain ; and
| | - Manuel Quintana
- Epilepsy Unit, Department of Neurology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Marilyn Riveiro
- Neurotrauma Intensive Care Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Juan Sahuquillo
- Neurotrauma and Neurosurgery Research Unit (UNINN), Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Department of Neurosurgery, Vall d'Hebron University Hospital, Barcelona, Spain ; and
| |
Collapse
|
22
|
Huang Y, Ni N, Hong Y, Lin X, Feng Y, Shen L. Progress in Traditional Chinese Medicine for the Treatment of Migraine. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1731-1748. [DOI: 10.1142/s0192415x2050086x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Migraine is a recurrent disease with complex pathogenesis and is difficult to cure. At present, commercially available western migraine drugs are prone to generate side effects while treating the disease. Traditional Chinese medicine (TCM) avoids side effects via treatment with the principles of “treating both symptoms and root causes”, “overall adjustment”, and “treatment based on syndrome differentiation”. Three strategies of drug treatment were developed based on the syndromes, i.e., removing stasis, calming liver Yang, and reinforcing deficiency. Prescriptions of removing stasis mostly contain Chuanxiong rhizome (Chuan Xiong) to remove blood stasis by promoting blood circulation and improve properties of hemorheology, and Da Chuan Xiong Formula (DCXF) is a traditional prescription widely used in clinical practice. Prescriptions of calming liver Yang usually take Ramulus Uncariae cum Uncis (Gou Teng) as the main herb, which can calm the liver Yang via improving vasomotor function, and Tian Ma Gou Teng Decoction (TMGTD) is the representative drug. For reinforcing deficiency, Chinese doctors frequently utilize Angelica Sinensis (Dang Gui) and Astragali Radix (Huang Qi) to nourish blood and Qi in order to improve the weak state of human body; Dang Gui Bu Xue Decoction (DGBXD) is the commonly used prescription. These strategies not only treat the symptoms of diseases but also their root causes, and with the features of multiple targets, in multiple ways. Therefore, TCM prescriptions have obvious advantages in the treatment of chronic diseases such as migraine. In this review, we provided an overview of the pathogenesis of migraine and the function of representative TCM preparations in therapy of migraine as well as the mechanism of action according to effective researches, in order to provide reference and clue for further researches.
Collapse
Affiliation(s)
- Yanleng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Ni Ni
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Yanlong Hong
- Health Service Collaborative Innovation Center of Shanghai, Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Xiao Lin
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Yi Feng
- Engineering Research Center of Modern Preparation Technology of Traditional Chinese Medicine of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| |
Collapse
|
23
|
Takizawa T, Qin T, Lopes de Morais A, Sugimoto K, Chung JY, Morsett L, Mulder I, Fischer P, Suzuki T, Anzabi M, Böhm M, Qu WS, Yanagisawa T, Hickman S, Khoury JE, Whalen MJ, Harriott AM, Chung DY, Ayata C. Non-invasively triggered spreading depolarizations induce a rapid pro-inflammatory response in cerebral cortex. J Cereb Blood Flow Metab 2020; 40:1117-1131. [PMID: 31242047 PMCID: PMC7181092 DOI: 10.1177/0271678x19859381] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cortical spreading depolarization (CSD) induces pro-inflammatory gene expression in brain tissue. However, previous studies assessing the relationship between CSD and inflammation have used invasive methods that directly trigger inflammation. To eliminate the injury confounder, we induced CSDs non-invasively through intact skull using optogenetics in Thy1-channelrhodopsin-2 transgenic mice. We corroborated our findings by minimally invasive KCl-induced CSDs through thinned skull. Six CSDs induced over 1 h dramatically increased cortical interleukin-1β (IL-1β), chemokine (C-C motif) ligand 2 (CCL2), and tumor necrosis factor-α (TNF-α) mRNA expression peaking around 1, 2 and 4 h, respectively. Interleukin-6 (IL-6) and intercellular adhesion molecule-1 (ICAM-1) were only modestly elevated. A single CSD also increased IL-1β, CCL2, and TNF-α, and revealed an ultra-early IL-1β response within 10 min. The response was blunted in IL-1 receptor-1 knockout mice, implicating IL-1β as an upstream mediator, and suppressed by dexamethasone, but not ibuprofen. CSD did not alter systemic inflammatory indices. In summary, this is the first report of pro-inflammatory gene expression after non-invasively induced CSDs. Altogether, our data provide novel insights into the role of CSD-induced neuroinflammation in migraine headache pathogenesis and have implications for the inflammatory processes in acute brain injury where numerous CSDs occur for days.
Collapse
Affiliation(s)
- Tsubasa Takizawa
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Tao Qin
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Andreia Lopes de Morais
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Kazutaka Sugimoto
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Joon Yong Chung
- Neuroscience Center, Massachusetts
General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Pediatrics, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liza Morsett
- Center for Immunology & Inflammatory
Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA,
USA
| | - Inge Mulder
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Paul Fischer
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Charité –
Universitätsmedizin Berlin, Berlin, Germany
| | - Tomoaki Suzuki
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Maryam Anzabi
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Maximilian Böhm
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Charité –
Universitätsmedizin Berlin, Berlin, Germany
| | - Wen-sheng Qu
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Takeshi Yanagisawa
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Suzanne Hickman
- Center for Immunology & Inflammatory
Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA,
USA
| | - Joseph El Khoury
- Center for Immunology & Inflammatory
Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA,
USA
| | - Michael J Whalen
- Neuroscience Center, Massachusetts
General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Pediatrics, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea M Harriott
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
| | - David Y Chung
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cenk Ayata
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
- Cenk Ayata, Massachusetts General Hospital,
149 13th Street, 6403, Charlestown, MA 02129, USA.
| |
Collapse
|
24
|
Ashayeri Ahmadabad R, Khaleghi Ghadiri M, Gorji A. The role of Toll-like receptor signaling pathways in cerebrovascular disorders: the impact of spreading depolarization. J Neuroinflammation 2020; 17:108. [PMID: 32264928 PMCID: PMC7140571 DOI: 10.1186/s12974-020-01785-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023] Open
Abstract
Cerebral vascular diseases (CVDs) are a group of disorders that affect the blood supply to the brain and lead to the reduction of oxygen and glucose supply to the neurons and the supporting cells. Spreading depolarization (SD), a propagating wave of neuroglial depolarization, occurs in different CVDs. A growing amount of evidence suggests that the inflammatory responses following hypoxic-ischemic insults and after SD plays a double-edged role in brain tissue injury and clinical outcome; a beneficial effect in the acute phase and a destructive role in the late phase. Toll-like receptors (TLRs) play a crucial role in the activation of inflammatory cascades and subsequent neuroprotective or harmful effects after CVDs and SD. Here, we review current data regarding the pathophysiological role of TLR signaling pathways in different CVDs and discuss the role of SD in the potentiation of the inflammatory cascade in CVDs through the modulation of TLRs.
Collapse
Affiliation(s)
- Rezan Ashayeri Ahmadabad
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Neuroscience research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
25
|
Dehghani A, Karatas H. Mouse Models of Familial Hemiplegic Migraine for Studying Migraine Pathophysiology. Curr Neuropharmacol 2020; 17:961-973. [PMID: 31092180 PMCID: PMC7052833 DOI: 10.2174/1570159x17666190513085013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/08/2019] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Migraine, an extremely disabling neurological disorder, has a strong genetic component. Since monogenic mi-graines (resulting from mutations or changes in a single gene) may help researchers discover migraine pathophysiology, transgenic mice models harboring gene mutations identified in Familial Hemiplegic Migraine (FHM) patients have been gen-erated. Studies in these FHM mutant mice models have shed light on the mechanisms of migraine and may aid in the identifi-cation of novel targets for treatment. More specifically, the studies shed light on how gene mutations, hormones, and other factors impact the pathophysiology of migraine. The models may also be of relevance to researchers outside the field of mi-graine as some of their aspects are relevant to pain in general. Additionally, because of the comorbidities associated with mi-graine, they share similarities with the mutant mouse models of epilepsy, stroke, and perhaps depression. Here, we review the experimental data obtained from these mutant mice and focus on how they can be used to investigate the pathophysiology of migraine, including synaptic plasticity, neuroinflammation, metabolite alterations, and molecular and behavioral mecha-nisms of pain.
Collapse
Affiliation(s)
- Anisa Dehghani
- Institute of Neurological Sciences and Psychiatry, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| |
Collapse
|
26
|
Tolner EA, Chen SP, Eikermann-Haerter K. Current understanding of cortical structure and function in migraine. Cephalalgia 2019; 39:1683-1699. [PMID: 30922081 PMCID: PMC6859601 DOI: 10.1177/0333102419840643] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To review and discuss the literature on the role of cortical structure and function in migraine. DISCUSSION Structural and functional findings suggest that changes in cortical morphology and function contribute to migraine susceptibility by modulating dynamic interactions across cortical and subcortical networks. The involvement of the cortex in migraine is well established for the aura phase with the underlying phenomenon of cortical spreading depolarization, while increasing evidence suggests an important role for the cortex in perception of head pain and associated sensations. As part of trigeminovascular pain and sensory processing networks, cortical dysfunction is likely to also affect initiation of attacks. CONCLUSION Morphological and functional changes identified across cortical regions are likely to contribute to initiation, cyclic recurrence and chronification of migraine. Future studies are needed to address underlying mechanisms, including interactions between cortical and subcortical regions and effects of internal (e.g. genetics, gender) and external (e.g. sensory inputs, stress) modifying factors, as well as possible clinical and therapeutic implications.
Collapse
Affiliation(s)
- Else A Tolner
- Departments of Neurology and Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Shih-Pin Chen
- Insitute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei
- Division of Translational Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei
| | | |
Collapse
|
27
|
Abstract
Migraine is among the most common and most disabling disorders worldwide, yet its underlying pathophysiology is among the most poorly understood. New information continues to emerge on mechanisms within the central and peripheral nervous systems that may contribute to migraine attacks. Additionally, new therapeutics have recently become available and along with much needed relief for many patients, these drugs provide insight into the disorder based on their mechanism of action. This review will cover new findings within the last several years that add to the understanding of migraine pathophysiology, including those related to the vasculature, calcitonin gene-related peptide (CGRP), and mechanisms within the cortex and meninges that may contribute to attacks. Discussion will also cover recent findings on novel therapeutic targets, several of which continue to show promise in new preclinical studies, including acid-sensing ion channels (ASICs) and the delta-opioid receptor (DOR).
Collapse
Affiliation(s)
- Greg Dussor
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080
| |
Collapse
|
28
|
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: 4.6] [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
|
29
|
Magni G, Boccazzi M, Bodini A, Abbracchio MP, van den Maagdenberg AMJM, Ceruti S. Basal astrocyte and microglia activation in the central nervous system of Familial Hemiplegic Migraine Type I mice. Cephalalgia 2019; 39:1809-1817. [DOI: 10.1177/0333102419861710] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Gain-of-function missense mutations in the α1A subunit of neuronal CaV2.1 channels, which define Familial Hemiplegic Migraine Type 1 (FHM1), result in enhanced cortical glutamatergic transmission and a higher susceptibility to cortical spreading depolarization. It is now well established that neurons signal to surrounding glial cells, namely astrocytes and microglia, in the central nervous system, which in turn become activated and in pathological conditions can sustain neuroinflammation. We and others previously demonstrated an increased activation of pro-algogenic pathways, paralleled by augmented macrophage infiltration, in both isolated trigeminal ganglia and mixed trigeminal ganglion neuron-satellite glial cell cultures of FHM1 mutant mice. Hence, we hypothesize that astrocyte and microglia activation may occur in parallel in the central nervous system. Methods We have evaluated signs of reactive glia in brains from naïve FHM1 mutant mice in comparison with wild type animals by immunohistochemistry and Western blotting. Results Here we show for the first time signs of reactive astrogliosis and microglia activation in the naïve FHM1 mutant mouse brain. Conclusions Our data reinforce the involvement of glial cells in migraine, and suggest that modulating such activation may represent an innovative approach to reduce pathology.
Collapse
Affiliation(s)
- Giulia Magni
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Marta Boccazzi
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Antonella Bodini
- Institute for Applied Mathematics and Information Technologies “Enrico Magenes”, National Research Council, Milan, Italy
| | - Maria P Abbracchio
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | | | - Stefania Ceruti
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
30
|
Zeng Z, Li Y, Lu S, Huang W, Di W. Efficacy of CoQ10 as supplementation for migraine: A meta-analysis. Acta Neurol Scand 2019; 139:284-293. [PMID: 30428123 DOI: 10.1111/ane.13051] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 10/10/2018] [Accepted: 10/27/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Migraine ranks among the most frequent neurological disorders globally. Co-enzyme Q10 (CoQ10) is a nutritional agent that might play a preventative role in migraine. This meta-analysis aimed to investigate the effects of CoQ10 as a supplemental agent in migraine. SUBJECTS AND METHODS Web of Science, PubMed, and Cochrane Library were searched for potential articles that assessed the effects of CoQ10 on migraine. Data were extracted by two independent reviewers and analyzed with Revman 5.2 software (The Nordic Cochrane Centre, Copenhagen, Denmark). RESULTS We included five studies with 346 patients (120 pediatric and 226 adult subjects) in the meta-analysis. CoQ10 was comparable with placebo with respect to migraine attacks/month (P = 0.08) and migraine severity/day (P = 0.08). However, CoQ10 was more effective than placebo in reducing migraine days/month (P < 0.00001) and migraine duration (P = 0.009). CONCLUSION This is the first study to demonstrate the effects of CoQ10 supplementation on migraine. The results support the use of CoQ10 as a potent therapeutic agent with respect to migraine duration and migraine days/month. Nonetheless, more studies are needed to support the conclusions.
Collapse
Affiliation(s)
- ZhiYong Zeng
- Department of Pediatrics; The Second People’s Hospital of Longgang District; Shenzhen China
| | - YunPeng Li
- Department of Neurosurgery; The People’s Hospital of Ningdu Country; Ganzhou China
| | - ShunYu Lu
- Department of Pharmacy; The Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker’s Hospital; Liuzhou China
| | - WanSu Huang
- Department of Criminal Science and Technology; Guangxi Police College; Nanning China
| | - Wei Di
- Department of Neurology; Shaanxi Provincial People’s Hospital, Third Affiliated Hospital of Medical College, Xi’an Jiaotong University; Xi’an China
| |
Collapse
|
31
|
Jeong H, Moye LS, Southey BR, Hernandez AG, Dripps I, Romanova EV, Rubakhin SS, Sweedler JV, Pradhan AA, Rodriguez-Zas SL. Gene Network Dysregulation in the Trigeminal Ganglia and Nucleus Accumbens of a Model of Chronic Migraine-Associated Hyperalgesia. Front Syst Neurosci 2018; 12:63. [PMID: 30618656 PMCID: PMC6305622 DOI: 10.3389/fnsys.2018.00063] [Citation(s) in RCA: 18] [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: 08/03/2018] [Accepted: 12/03/2018] [Indexed: 12/28/2022] Open
Abstract
The pharmacological agent nitroglycerin (NTG) elicits hyperalgesia and allodynia in mice. This model has been used to study the neurological disorder of trigeminovascular pain or migraine, a debilitating form of hyperalgesia. The present study validates hyperalgesia in an established mouse model of chronic migraine triggered by NTG and advances the understanding of the associated molecular mechanisms. The RNA-seq profiles of two nervous system regions associated with pain, the trigeminal ganglia (TG) and the nucleus accumbens (NAc), were compared in mice receiving chronic NTG treatment relative to control (CON) mice. Among the 109 genes that exhibited an NTG treatment-by-region interaction, solute carrier family 32 (GABA vesicular transporter) member 1 (Slc32a1) and preproenkephalin (Penk) exhibited reversal of expression patterns between the NTG and CON groups. Erb-b2 receptor tyrosine kinase 4 (Erbb4) and solute carrier family 1 (glial high affinity glutamate transporter) member 2 (Slc1a2) exhibited consistent differential expression between treatments across regions albeit at different magnitude. Period circadian clock 1 (Per1) was among the 165 genes that exhibited significant NTG treatment effect. Biological processes disrupted by NTG in a region-specific manner included adaptive and innate immune responses; whereas glutamatergic and dopaminergic synapses and rhythmic process were disrupted in both regions. Regulatory network reconstruction highlighted the widespread role of several transcription factors (including Snrnp70, Smad1, Pax6, Cebpa, and Smpx) among the NTG-disrupted target genes. These results advance the understanding of the molecular mechanisms of hyperalgesia that can be applied to therapies to ameliorate chronic pain and migraine.
Collapse
Affiliation(s)
- Hyeonsoo Jeong
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Laura S. Moye
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Bruce R. Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Alvaro G. Hernandez
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Isaac Dripps
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Elena V. Romanova
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Stanislav S. Rubakhin
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Jonathan V. Sweedler
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Amynah A. Pradhan
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Sandra L. Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
32
|
Klass A, Sánchez-Porras R, Santos E. Systematic review of the pharmacological agents that have been tested against spreading depolarizations. J Cereb Blood Flow Metab 2018; 38:1149-1179. [PMID: 29673289 PMCID: PMC6434447 DOI: 10.1177/0271678x18771440] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Spreading depolarization (SD) occurs alongside brain injuries and it can lead to neuronal damage. Therefore, pharmacological modulation of SD can constitute a therapeutic approach to reduce its detrimental effects and to improve the clinical outcome of patients. The major objective of this article was to produce a systematic review of all the drugs that have been tested against SD. Of the substances that have been examined, most have been shown to modulate certain SD characteristics. Only a few have succeeded in significantly inhibiting SD. We present a variety of strategies that have been proposed to overcome the notorious harmfulness and pharmacoresistance of SD. Information on clinically used anesthetic, sedative, hypnotic agents, anti-migraine drugs, anticonvulsants and various other substances have been compiled and reviewed with respect to the efficacy against SD, in order to answer the question of whether a drug at safe doses could be of therapeutic use against SD in humans.
Collapse
Affiliation(s)
- Anna Klass
- Neurosurgery Department, University of Heidelberg, Heidelberg, Germany
| | | | - Edgar Santos
- Neurosurgery Department, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
33
|
Cozzolino O, Marchese M, Trovato F, Pracucci E, Ratto GM, Buzzi MG, Sicca F, Santorelli FM. Understanding Spreading Depression from Headache to Sudden Unexpected Death. Front Neurol 2018; 9:19. [PMID: 29449828 PMCID: PMC5799941 DOI: 10.3389/fneur.2018.00019] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/11/2018] [Indexed: 01/03/2023] Open
Abstract
Spreading depression (SD) is a neurophysiological phenomenon characterized by abrupt changes in intracellular ion gradients and sustained depolarization of neurons. It leads to loss of electrical activity, changes in the synaptic architecture, and an altered vascular response. Although SD is often described as a unique phenomenon with homogeneous characteristics, it may be strongly affected by the particular triggering event and by genetic background. Furthermore, SD may contribute differently to the pathogenesis of widely heterogeneous clinical conditions. Indeed, clinical disorders related to SD vary in their presentation and severity, ranging from benign headache conditions (migraine syndromes) to severely disabling events, such as cerebral ischemia, or even death in people with epilepsy. Although the characteristics and mechanisms of SD have been dissected using a variety of approaches, ranging from cells to human models, this phenomenon remains only partially understood because of its complexity and the difficulty of obtaining direct experimental data. Currently, clinical monitoring of SD is limited to patients who require neurosurgical interventions and the placement of subdural electrode strips. Significantly, SD events recorded in humans display electrophysiological features that are essentially the same as those observed in animal models. Further research using existing and new experimental models of SD may allow a better understanding of its core mechanisms, and of their differences in different clinical conditions, fostering opportunities to identify and develop targeted therapies for SD-related disorders and their worst consequences.
Collapse
Affiliation(s)
- Olga Cozzolino
- NEST, Istituto Nanoscienze CNR and Scuola Normale Superiore, Pisa, Italy
| | - Maria Marchese
- Molecular Medicine and Clinical Neurophysiology Laboratories, Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Francesco Trovato
- NEST, Istituto Nanoscienze CNR and Scuola Normale Superiore, Pisa, Italy
| | - Enrico Pracucci
- NEST, Istituto Nanoscienze CNR and Scuola Normale Superiore, Pisa, Italy
| | - Gian Michele Ratto
- NEST, Istituto Nanoscienze CNR and Scuola Normale Superiore, Pisa, Italy
| | | | - Federico Sicca
- Molecular Medicine and Clinical Neurophysiology Laboratories, Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Filippo M Santorelli
- Molecular Medicine and Clinical Neurophysiology Laboratories, Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa, Italy
| |
Collapse
|
34
|
Chen SP, Qin T, Seidel JL, Zheng Y, Eikermann M, Ferrari MD, van den Maagdenberg AMJM, Moskowitz MA, Ayata C, Eikermann-Haerter K. Inhibition of the P2X7-PANX1 complex suppresses spreading depolarization and neuroinflammation. Brain 2017; 140:1643-1656. [PMID: 28430869 DOI: 10.1093/brain/awx085] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 02/12/2017] [Indexed: 01/09/2023] Open
Abstract
Spreading depolarization is a wave of neuronal and glial depolarization. Within minutes after spreading depolarization, the neuronal hemichannel pannexin 1 (PANX1) opens and forms a pore complex with the ligand-gated cation channel P2X7, allowing the release of excitatory neurotransmitters to sustain spreading depolarization and activate neuroinflammation. Here, we explore the hypothesis that the P2X7-PANX1 pore complex is a critical determinant of spreading depolarization susceptibility with important consequences for neuroinflammation and trigeminovascular activation. We found that genetic loss of function or ablation of the P2x7 gene inhibits spreading depolarization. Moreover, pharmacological suppression of the P2X7-PANX1 pore complex inhibits spreading depolarization in mice carrying the human familial hemiplegic migraine type 1 R192Q missense mutation as well as in wild-type mice and rats. Pore inhibitors elevate the electrical threshold for spreading depolarization, and reduce spreading depolarization frequency and amplitude. Pore inhibitors also suppress downstream consequences of spreading depolarization such as upregulation of interleukin-1 beta, inducible nitric oxide synthase and cyclooxygenase-2 in the cortex after spreading depolarization. In addition, they inhibit surrogates for trigeminovascular activation, including expression of calcitonin gene-related peptide in the trigeminal ganglion and c-Fos in the trigeminal nucleus caudalis. Our results are consistent with the hypothesis that the P2X7-PANX1 pore complex is a critical determinant of spreading depolarization susceptibility and its downstream consequences, of potential relevance to its signature disorders such as migraine.
Collapse
Affiliation(s)
- Shih-Pin Chen
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Tao Qin
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Jessica L Seidel
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yi Zheng
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Matthias Eikermann
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA and Universitaet Duisburg Essen, Essen, Germany
| | - Michel D Ferrari
- Department of Neurology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Neurology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | - Michael A Moskowitz
- Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Cenk Ayata
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Katharina Eikermann-Haerter
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
35
|
Sutherland HG, Griffiths LR. Genetics of Migraine: Insights into the Molecular Basis of Migraine Disorders. Headache 2017; 57:537-569. [PMID: 28271496 DOI: 10.1111/head.13053] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/09/2017] [Indexed: 12/20/2022]
Abstract
Migraine is a complex, debilitating neurovascular disorder, typically characterized by recurring, incapacitating attacks of severe headache often accompanied by nausea and neurological disturbances. It has a strong genetic basis demonstrated by rare migraine disorders caused by mutations in single genes (monogenic), as well as familial clustering of common migraine which is associated with polymorphisms in many genes (polygenic). Hemiplegic migraine is a dominantly inherited, severe form of migraine with associated motor weakness. Family studies have found that mutations in three different ion channels genes, CACNA1A, ATP1A2, and SCN1A can be causal. Functional studies of these mutations has shown that they can result in defective regulation of glutamatergic neurotransmission and the excitatory/inhibitory balance in the brain, which lowers the threshold for cortical spreading depression, a wave of cortical depolarization thought to be involved in headache initiation mechanisms. Other putative genes for monogenic migraine include KCKN18, PRRT2, and CSNK1D, which can also be involved with other disorders. There are a number of primarily vascular disorders caused by mutations in single genes, which are often accompanied by migraine symptoms. Mutations in NOTCH3 causes cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a hereditary cerebrovascular disease that leads to ischemic strokes and dementia, but in which migraine is often present, sometimes long before the onset of other symptoms. Mutations in the TREX1 and COL4A1 also cause vascular disorders, but often feature migraine. With respect to common polygenic migraine, genome-wide association studies have now identified single nucleotide polymorphisms at 38 loci significantly associated with migraine risk. Functions assigned to the genes in proximity to these loci suggest that both neuronal and vascular pathways also contribute to the pathophysiology of common migraine. Further studies are required to fully understand these findings and translate them into treatment options for migraine patients.
Collapse
Affiliation(s)
- Heidi G Sutherland
- Genomics Research Centre, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, QUT, Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, QUT, Musk Ave, Kelvin Grove, QLD, 4059, Australia
| |
Collapse
|
36
|
Gasparini CF, Smith RA, Griffiths LR. Genetic insights into migraine and glutamate: a protagonist driving the headache. J Neurol Sci 2016; 367:258-68. [PMID: 27423601 DOI: 10.1016/j.jns.2016.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 05/11/2016] [Accepted: 06/08/2016] [Indexed: 12/12/2022]
Abstract
Migraine is a complex polygenic disorder that continues to be a great source of morbidity in the developed world with a prevalence of 12% in the Caucasian population. Genetic and pharmacological studies have implicated the glutamate pathway in migraine pathophysiology. Glutamate profoundly impacts brain circuits that regulate core symptom domains in a range of neuropsychiatric conditions and thus remains a "hot" target for drug discovery. Glutamate has been implicated in cortical spreading depression (CSD), the phenomenon responsible for migraine with aura and in animal models carrying FHM mutations. Genotyping case-control studies have shown an association between glutamate receptor genes, namely, GRIA1 and GRIA3 with migraine with indirect supporting evidence from GWAS. New evidence localizes PRRT2 at glutamatergic synapses and shows it affects glutamate signalling and glutamate receptor activity via interactions with GRIA1. Glutamate-system defects have also been recently implicated in a novel FHM2 ATP1A2 disease-mutation mouse model. Adding to the growing evidence neurophysiological findings support a role for glutamate in cortical excitability. In addition to the existence of multiple genes to choreograph the functions of fast-signalling glutamatergic neurons, glutamate receptor diversity and regulation is further increased by the post-translational mechanisms of RNA editing and miRNAs. Ongoing genetic studies, GWAS and meta-analysis implicate neurogenic mechanisms in migraine pathology and the first genome-wide associated locus for migraine on chromosome X. Finally, in addition to glutamate modulating therapies, the kynurenine pathway has emerged as a candidate for involvement in migraine pathophysiology. In this review we discuss recent genetic evidence and glutamate modulating therapies that bear on the hypothesis that a glutamatergic mechanism may be involved in migraine susceptibility.
Collapse
Affiliation(s)
- Claudia F Gasparini
- Menzies Health Institute Queensland, Griffith University Gold Coast, Parklands Drive, Southport, QLD 4222, Australia
| | - Robert A Smith
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Musk Ave, Kelvin Grove, QLD 4059, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Musk Ave, Kelvin Grove, QLD 4059, Australia.
| |
Collapse
|
37
|
Kwong KK, Chan ST. Neuroprotection and acidosis induced by cortical spreading depression. Neuropsychiatr Dis Treat 2016; 12:3191-3194. [PMID: 28003755 PMCID: PMC5161389 DOI: 10.2147/ndt.s125677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Kenneth K Kwong
- Department of Radiology, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Suk-Tak Chan
- Department of Radiology, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|