51
|
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: 121] [Impact Index Per Article: 30.3] [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
|
52
|
Inoue H, Kuroda H, Ofusa W, Oyama S, Kimura M, Ichinohe T, Shibukawa Y. Functional Coupling between the P2X 7 Receptor and Pannexin-1 Channel in Rat Trigeminal Ganglion Neurons. Int J Mol Sci 2021; 22:ijms22115978. [PMID: 34205953 PMCID: PMC8198496 DOI: 10.3390/ijms22115978] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
The ionotropic P2X receptor, P2X7, is believed to regulate and/or generate nociceptive pain, and pain in several neuropathological diseases. Although there is a known relationship between P2X7 receptor activity and pain sensing, its detailed functional properties in trigeminal ganglion (TG) neurons remains unclear. We examined the electrophysiological and pharmacological characteristics of the P2X7 receptor and its functional coupling with other P2X receptors and pannexin-1 (PANX1) channels in primary cultured rat TG neurons, using whole-cell patch-clamp recordings. Application of ATP and Bz-ATP induced long-lasting biphasic inward currents that were more sensitive to extracellular Bz-ATP than ATP, indicating that the current was carried by P2X7 receptors. While the biphasic current densities of the first and second components were increased by Bz-ATP in a concentration dependent manner; current duration was only affected in the second component. These currents were significantly inhibited by P2X7 receptor antagonists, while only the second component was inhibited by P2X1, 3, and 4 receptor antagonists, PANX1 channel inhibitors, and extracellular ATPase. Taken together, our data suggests that autocrine or paracrine signaling via the P2X7-PANX1-P2X receptor/channel complex may play important roles in several pain sensing pathways via long-lasting neuronal activity driven by extracellular high-concentration ATP following tissue damage in the orofacial area.
Collapse
Affiliation(s)
- Hiroyuki Inoue
- Department of Physiology, Tokyo Dental College, Tokyo 101-0061, Japan; (H.I.); (H.K.); (W.O.); (S.O.); (M.K.)
- Department of Dental Anesthesiology, Tokyo Dental College, Tokyo 101-0061, Japan;
| | - Hidetaka Kuroda
- Department of Physiology, Tokyo Dental College, Tokyo 101-0061, Japan; (H.I.); (H.K.); (W.O.); (S.O.); (M.K.)
- Department of Dental Anesthesiology, Kanagawa Dental University, Kanagawa 238-8580, Japan
| | - Wataru Ofusa
- Department of Physiology, Tokyo Dental College, Tokyo 101-0061, Japan; (H.I.); (H.K.); (W.O.); (S.O.); (M.K.)
| | - Sadao Oyama
- Department of Physiology, Tokyo Dental College, Tokyo 101-0061, Japan; (H.I.); (H.K.); (W.O.); (S.O.); (M.K.)
| | - Maki Kimura
- Department of Physiology, Tokyo Dental College, Tokyo 101-0061, Japan; (H.I.); (H.K.); (W.O.); (S.O.); (M.K.)
| | - Tatsuya Ichinohe
- Department of Dental Anesthesiology, Tokyo Dental College, Tokyo 101-0061, Japan;
| | - Yoshiyuki Shibukawa
- Department of Physiology, Tokyo Dental College, Tokyo 101-0061, Japan; (H.I.); (H.K.); (W.O.); (S.O.); (M.K.)
- Correspondence:
| |
Collapse
|
53
|
Zeng Y, Luo H, Gao Z, Zhu X, Shen Y, Li Y, Hu J, Yang J. Reduction of prefrontal purinergic signaling is necessary for the analgesic effect of morphine. iScience 2021; 24:102213. [PMID: 33733073 PMCID: PMC7940985 DOI: 10.1016/j.isci.2021.102213] [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: 11/23/2020] [Revised: 01/26/2021] [Accepted: 02/17/2021] [Indexed: 01/14/2023] Open
Abstract
Morphine is commonly used to relieve moderate to severe pain, but repeated doses cause opioid tolerance. Here, we used ATP sensor and fiber photometry to detect prefrontal ATP level. It showed that prefrontal ATP level decreased after morphine injection and the event amplitude tended to decrease with continuous morphine exposure. Morphine had little effect on prefrontal ATP due to its tolerance. Therefore, we hypothesized that the analgesic effect of morphine might be related to ATP in the medial prefrontal cortex (mPFC). Moreover, local infusion of ATP partially antagonized morphine analgesia. Then we found that inhibiting P2X7R in the mPFC mimicked morphine analgesia. In morphine-tolerant mice, pretreatment with P2X4R or P2X7R antagonists in the mPFC enhanced analgesic effect. Our findings suggest that reduction of prefrontal purinergic signaling is necessary for the morphine analgesia, which help elucidate the mechanism of morphine analgesia and may lead to the development of new clinical treatments for neuropathic pain.
Collapse
Affiliation(s)
- Yeting Zeng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zilong Gao
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing 102206, China
| | - Xiaona Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yinbo Shen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yulong Li
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing 102206, China
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, 200030, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
- gCAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200030, China
| | - Jiajun Yang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
54
|
Welcome MO, Mastorakis NE. The taste of neuroinflammation: Molecular mechanisms linking taste sensing to neuroinflammatory responses. Pharmacol Res 2021; 167:105557. [PMID: 33737243 DOI: 10.1016/j.phrs.2021.105557] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
Evidence indicates a critical role of neuroinflammatory response as an underlying pathophysiological process in several central nervous system disorders, including neurodegenerative diseases. However, the molecular mechanisms that trigger neuroinflammatory processes are not fully known. The discovery of bitter taste receptors in regions other than the oral cavity substantially increased research interests on their functional roles in extra-oral tissues. It is now widely accepted that bitter taste receptors, for instance, in the respiratory, intestinal, reproductive and urinary tracts, are crucial not only for sensing poisonous substances, but also, act as immune sentinels, mobilizing defense mechanisms against pathogenic aggression. The relatively recent discovery of bitter taste receptors in the brain has intensified research investigation on the functional implication of cerebral bitter taste receptor expression. Very recent data suggest that responses of bitter taste receptors to neurotoxins and microbial molecules, under normal condition, are necessary to prevent neuroinflammatory reactions. Furthermore, emerging data have revealed that downregulation of key components of the taste receptor signaling cascade leads to increased oxidative stress and inflammasome signaling in neurons that ultimately culminate in neuroinflammation. Nevertheless, the mechanisms that link taste receptor mediated surveillance of the extracellular milieu to neuroinflammatory responses are not completely understood. This review integrates new data on the molecular mechanisms that link bitter taste receptor sensing to neuroinflammatory responses. The role of bitter taste receptor-mediated sensing of toxigenic substances in brain disorders is also discussed. The therapeutic significance of targeting these receptors for potential treatment of neurodegenerative diseases is also highlighted.
Collapse
Affiliation(s)
- Menizibeya O Welcome
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, Abuja, Nigeria.
| | | |
Collapse
|
55
|
Eikermann-Haerter K. Neuronal plumes initiate spreading depolarization, the electrophysiologic event driving migraine and stroke. Neuron 2021; 109:563-565. [PMID: 33600751 DOI: 10.1016/j.neuron.2021.01.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this issue of Neuron, Parker et al. discover neuronal plumes of glutamate release that initiate spreading depolarization, the electrophysiologic event underlying migraine. Mice with human migraine mutations express spontaneous and frequent plumes, which may explain the propensity to develop migraine attacks and the increased stroke risk in migraine-susceptible brains.
Collapse
|
56
|
Eikermann-Haerter K, Huang SY. White Matter Lesions in Migraine. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1955-1962. [PMID: 33636178 DOI: 10.1016/j.ajpath.2021.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/16/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022]
Abstract
Migraine, the third most common disease worldwide, is a well-known independent risk factor for subclinical focal deep white matter lesions (WMLs), even in young and otherwise healthy individuals with no cardiovascular risk factors. These WMLs are more commonly seen in migraine patients with transient neurologic symptoms preceding their headaches, the so-called aura, and those with a high attack frequency. The pathophysiology of migraine-related deep white matter hyperintensities remains poorly understood despite their prevalence. Characteristic differences in their distribution related to chronic small vessel ischemic disease compared with that of common periventricular WMLs in the elderly suggest a different underlying mechanism. Both ischemic and inflammatory mechanisms have been proposed, as there is increased cerebral vulnerability to ischemia in migraineurs, whereas there is also evidence of blood-brain barrier disruption with associated release of proinflammatory substances during migraine attacks. An enhanced susceptibility to spreading depolarization, the electrophysiological event underlying migraine, may be the mechanism that causes repetitive episodes of cerebral hypoperfusion and neuroinflammation during migraine attacks. WMLs can negatively affect both physical and cognitive function, underscoring the public health importance of migraine, and suggesting that migraine is an important contributor to neurologic deficits in the general population.
Collapse
Affiliation(s)
| | - Susie Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and the Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
57
|
Szopa A, Socała K, Serefko A, Doboszewska U, Wróbel A, Poleszak E, Wlaź P. Purinergic transmission in depressive disorders. Pharmacol Ther 2021; 224:107821. [PMID: 33607148 DOI: 10.1016/j.pharmthera.2021.107821] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Purinergic signaling involves the actions of purine nucleotides and nucleosides (such as adenosine) at P1 (adenosine), P2X, and P2Y receptors. Here, we present recent data contributing to a comprehensive overview of the association between purinergic signaling and depression. We start with background information on adenosine production and metabolism, followed by a detailed characterization of P1 and P2 receptors, with an emphasis on their expression and function in the brain as well as on their ligands. We provide data suggestive of altered metabolism of adenosine in depressed patients, which might be regarded as a disease biomarker. We then turn to considerable amount of preclinical/behavioral data obtained with the aid of the forced swim test, tail suspension test, learned helplessness model, or unpredictable chronic mild stress model and genetic activation/inactivation of P1 or P2 receptors as well as nonselective or selective ligands of P1 or P2 receptors. We also aimed to discuss the reason underlying discrepancies observed in such studies.
Collapse
Affiliation(s)
- Aleksandra Szopa
- Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, PL 20-093 Lublin, Poland.
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland
| | - Anna Serefko
- Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, PL 20-093 Lublin, Poland
| | - Urszula Doboszewska
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland
| | - Andrzej Wróbel
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego 8, PL 20-090 Lublin, Poland
| | - Ewa Poleszak
- Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, PL 20-093 Lublin, Poland.
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland.
| |
Collapse
|
58
|
Petit JM, Eren-Koçak E, Karatas H, Magistretti P, Dalkara T. Brain glycogen metabolism: A possible link between sleep disturbances, headache and depression. Sleep Med Rev 2021; 59:101449. [PMID: 33618186 DOI: 10.1016/j.smrv.2021.101449] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/27/2022]
Abstract
The functions of sleep and its links with neuropsychiatric diseases have long been questioned. Among the numerous hypotheses on sleep function, early studies proposed that sleep helps to replenish glycogen stores consumed during waking. Later studies found increased brain glycogen after sleep deprivation, leading to "glycogenetic" hypothesis, which states that there is a parallel increase in synthesis and utilization of glycogen during wakefulness, whereas decrease in the excitatory transmission creates an imbalance causing accumulation of glycogen during sleep. Glycogen is a vital energy reservoir to match the synaptic demand particularly for re-uptake of potassium and glutamate during intense glutamatergic transmission. Therefore, sleep deprivation-induced transcriptional changes may trigger migraine by reducing glycogen availability, which slows clearance of extracellular potassium and glutamate, hence, creates susceptibility to cortical spreading depolarization, the electrophysiological correlate of migraine aura. Interestingly, chronic stress accompanied by increased glucocorticoid levels and locus coeruleus activity and leading to mood disorders in which sleep disturbances are prevalent, also affects brain glycogen turnover via glucocorticoids, noradrenaline, serotonin and adenosine. These observations altogether suggest that inadequate astrocytic glycogen turnover may be one of the mechanisms linking migraine, mood disorders and sleep.
Collapse
Affiliation(s)
- J-M Petit
- Lausanne University Hospital, Center for Psychiatric Neuroscience, Prilly, Switzerland.
| | - E Eren-Koçak
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, and Faculty of Medicine, Department of Psychiatry, Ankara, Turkey.
| | - H Karatas
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey.
| | - P Magistretti
- King Abdullah University of Science and Technology, Saudi Arabia.
| | - T Dalkara
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey.
| |
Collapse
|
59
|
Homozygous variants in PANX1 cause human oocyte death and female infertility. Eur J Hum Genet 2021; 29:1396-1404. [PMID: 33495594 DOI: 10.1038/s41431-020-00807-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/20/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
PANX1, one of the members of the pannexin family, is a highly glycosylated channel-forming protein. Recently, we identified heterozygous variants in PANX1 that follow an autosomal dominant inheritance pattern and cause female infertility characterized by oocyte death. In this study, we screened for novel PANX1 variants in patients with the phenotype of oocyte death and discovered a new type of inheritance pattern accompanying PANX1 variants. We identified two novel homozygous missense variants in PANX1 [NM_015368.4 c.712T>C (p.(Ser238Pro) and c.899G>A (p.(Arg300Gln))] associated with the oocyte death phenotype in two families. Both of the homozygous variants altered the PANX1 glycosylation pattern in cultured cells, led to aberrant PANX1 channel activation, and resulted in mouse oocyte death after fertilization in vitro. It is worth noting that the destructive effect of the two homozygous variants on PANX1 function was weaker than that caused by the recently reported heterozygous variants. Our findings enrich the variational spectrum of PANX1 and expand the inheritance pattern of PANX1 variants to an autosomal recessive mode. This highlights the critical role of PANX1 in human oocyte development and helps us to better understand the genetic basis of female infertility due to oocyte death.
Collapse
|
60
|
Pusic KM, Won L, Kraig RP, Pusic AD. Environmental Enrichment and Its Benefits for Migraine: Dendritic Cell Extracellular Vesicles as an Effective Mimetic. JOURNAL OF CELLULAR IMMUNOLOGY 2021; 3:215-225. [PMID: 34337600 PMCID: PMC8321388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Environmental enrichment produces beneficial effects in the brain at genetic, molecular, cellular and behavior levels, and has long been studied as a therapeutic intervention for a wide variety of neurological disorders. However, the complexity of applying a robust environmental enrichment paradigm makes clinical use difficult. Accordingly, there has been increased interest in developing environmental enrichment mimetics, also known as enviromimetics. Here we review the benefits of environmental enrichment for migraine treatment, and discuss the potential of using extracellular vesicles derived from interferon gamma-stimulated dendritic cells as an effective mimetic.
Collapse
Affiliation(s)
- Kae Myriam Pusic
- Department of Neurology, The University of Chicago, Chicago, Il 60637-1470, USA
| | - Lisa Won
- Department of Neurology, The University of Chicago, Chicago, Il 60637-1470, USA
| | - Richard Paul Kraig
- Department of Neurology, The University of Chicago, Chicago, Il 60637-1470, USA
| | - Aya Darinka Pusic
- Department of Neurology, The University of Chicago, Chicago, Il 60637-1470, USA
| |
Collapse
|
61
|
Varga DP, Menyhárt Á, Pósfai B, Császár E, Lénárt N, Cserép C, Orsolits B, Martinecz B, Szlepák T, Bari F, Farkas E, Dénes Á. Microglia alter the threshold of spreading depolarization and related potassium uptake in the mouse brain. J Cereb Blood Flow Metab 2020; 40:S67-S80. [PMID: 31987008 PMCID: PMC7687034 DOI: 10.1177/0271678x19900097] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Selective elimination of microglia from the brain was shown to dysregulate neuronal Ca2+ signaling and to reduce the incidence of spreading depolarization (SD) during cerebral ischemia. However, the mechanisms through which microglia interfere with SD remained unexplored. Here, we identify microglia as essential modulators of the induction and evolution of SD in the physiologically intact brain in vivo. Confocal- and super-resolution microscopy revealed that a series of SDs induced rapid morphological changes in microglia, facilitated microglial process recruitment to neurons and increased the density of P2Y12 receptors (P2Y12R) on recruited microglial processes. In line with this, depolarization and hyperpolarization during SD were microglia- and P2Y12R-dependent. An absence of microglia was associated with altered potassium uptake after SD and increased the number of c-fos-positive neurons, independently of P2Y12R. Thus, the presence of microglia is likely to be essential to maintain the electrical elicitation threshold and to support the full evolution of SD, conceivably by interfering with the extracellular potassium homeostasis of the brain through sustaining [K+]e re-uptake mechanisms.
Collapse
Affiliation(s)
- Dániel P Varga
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Ákos Menyhárt
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Balázs Pósfai
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.,Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest, Hungary
| | - Eszter Császár
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.,Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest, Hungary
| | - Nikolett Lénárt
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Csaba Cserép
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Barbara Orsolits
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Bernadett Martinecz
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Tamás Szlepák
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.,Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest, Hungary
| | - Ferenc Bari
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Ádám Dénes
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
62
|
Currò D, Navarra P, Samengo I, Martire M. P2X7 receptors exert a permissive effect on the activation of presynaptic AMPA receptors in rat trigeminal caudal nucleus glutamatergic nerve terminals. J Headache Pain 2020; 21:83. [PMID: 32615921 PMCID: PMC7330953 DOI: 10.1186/s10194-020-01153-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background Purine receptors play roles in peripheral and central sensitization and are associated with migraine headache. We investigated the possibility that ATP plays a permissive role in the activation of AMPA receptors thus inducing Glu release from nerve terminals isolated from the rat trigeminal caudal nucleus (TCN). Methods Nerve endings isolated from the rat TCN were loaded with [3H]D-aspartic acid ([3H]D-ASP), layered into thermostated superfusion chambers, and perfused continuously with physiological medium, alone or with various test drugs. Radioactivity was measured to assess [3H]D-ASP release under different experimental conditions. Results Synaptosomal [3H]D-ASP spontaneous release was stimulated by ATP and to an even greater extent by the ATP analogue benzoylbenzoylATP (BzATP). The stimulation of [3H]D-ASP basal release by the purinergic agonists was prevented by the selective P2X7 receptor antagonist A438079. AMPA had no effect on basal [3H]D-ASP release, but the release observed when synaptosomes were exposed to AMPA plus a purinoceptor agonist exceeded that observed with ATP or BzATP alone. The selective AMPA receptor antagonist NBQX blocked this “excess” release. Co-exposure to AMPA and BzATP, each at a concentration with no release-stimulating effects, evoked a significant increase in [3H]D-ASP basal release, which was prevented by exposure to a selective AMPA antagonist. Conclusions P2X7 receptors expressed on glutamatergic nerve terminals in the rat TCN can mediate Glu release directly and indirectly by facilitating the activation of presynaptic AMPA receptors. The high level of glial ATP that occurs during chronic pain states can promote widespread release of Glu as well as can increase the function of AMPA receptors. In this manner, ATP contributes to the AMPA receptor activation involved in the onset and maintenance of the central sensitization associated with chronic pain.
Collapse
Affiliation(s)
- Diego Currò
- Institute of Pharmacology, School of Medicine, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Rome, Italy
| | - Pierluigi Navarra
- Institute of Pharmacology, School of Medicine, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Rome, Italy
| | - Irene Samengo
- Institute of Pharmacology, School of Medicine, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Rome, Italy
| | - Maria Martire
- Institute of Pharmacology, School of Medicine, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Rome, Italy.
| |
Collapse
|
63
|
Takizawa T, Ayata C, Chen SP. Therapeutic implications of cortical spreading depression models in migraine. PROGRESS IN BRAIN RESEARCH 2020; 255:29-67. [PMID: 33008510 DOI: 10.1016/bs.pbr.2020.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023]
Abstract
Migraine is among the most common and disabling neurological diseases in the world. Cortical spreading depression (CSD) is a wave of near-complete depolarization of neurons and glial cells that slowly propagates along the cortex creating the perception of aura. Evidence suggests that CSD can trigger migraine headache. Experimental models of CSD have been considered highly translational as they recapitulate migraine-related phenomena and have been validated for screening migraine therapeutics. Here we outline the essential components of validated experimental models of CSD and provide a comprehensive review of potential modulators and targets against CSD. We further focus on novel interventions that have been recently shown to suppress CSD susceptibility that may lead to therapeutic targets in migraine.
Collapse
Affiliation(s)
- Tsubasa Takizawa
- Department of Neurology, Keio Universrity School of Medicine, Tokyo, Japan
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States; Stroke Service, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Shih-Pin Chen
- Department of Medical Research & Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan; Brain Research Center, National Yang-Ming University School of Medicine, Taipei, Taiwan.
| |
Collapse
|
64
|
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: 11.2] [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
|
65
|
Argon Inhalation for 24 Hours After Onset of Permanent Focal Cerebral Ischemia in Rats Provides Neuroprotection and Improves Neurologic Outcome. Crit Care Med 2020; 47:e693-e699. [PMID: 31094741 DOI: 10.1097/ccm.0000000000003809] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES We tested the hypothesis that prolonged inhalation of 70% argon for 24 hours after in vivo permanent or temporary stroke provides neuroprotection and improves neurologic outcome and overall recovery after 7 days. DESIGN Controlled, randomized, double-blinded laboratory study. SETTING Animal research laboratories. SUBJECTS Adult Wistar male rats (n = 110). INTERVENTIONS Rats were subjected to permanent or temporary focal cerebral ischemia via middle cerebral artery occlusion, followed by inhalation of 70% argon or nitrogen in 30% oxygen for 24 hours. On postoperative day 7, a 48-point neuroscore and histologic lesion size were assessed. MEASUREMENTS AND MAIN RESULTS After argon inhalation for 24 hours immediately following "severe permanent ischemia" induction, neurologic outcome (neuroscore, p = 0.034), overall recovery (body weight, p = 0.02), and infarct volume (total infarct volume, p = 0.0001; cortical infarct volume, p = 0.0003; subcortical infarct volume, p = 0.0001) were significantly improved. When 24-hour argon treatment was delayed for 2 hours after permanent stroke induction or until after postischemic reperfusion treatment, neurologic outcomes remained significantly improved (neuroscore, p = 0.043 and p = 0.014, respectively), as was overall recovery (body weight, p = 0.015), compared with nitrogen treatment. However, infarct volume and 7-day mortality were not significantly reduced when argon treatment was delayed. CONCLUSIONS Neurologic outcome (neuroscore), overall recovery (body weight), and infarct volumes were significantly improved after 24-hour inhalation of 70% argon administered immediately after severe permanent stroke induction. Neurologic outcome and overall recovery were also significantly improved even when argon treatment was delayed for 2 hours or until after reperfusion.
Collapse
|
66
|
Stevens RD, Koehler RC. Pathophysiological Insights into Spreading Depolarization in Severe Traumatic Brain Injury. Neurocrit Care 2020; 30:569-571. [PMID: 30877553 DOI: 10.1007/s12028-019-00705-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Robert D Stevens
- Division of Neurosciences Critical Care, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Phipps 455, Baltimore, MD, 21287, USA. .,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
67
|
Zhang WJ, Zhu ZM, Liu ZX. The role and pharmacological properties of the P2X7 receptor in neuropathic pain. Brain Res Bull 2020; 155:19-28. [PMID: 31778766 DOI: 10.1016/j.brainresbull.2019.11.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/03/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Neuropathic Pain (NPP) is caused by direct or indirect damage to the nervous system and is a common symptom of many diseases. Clinically, drugs are usually used to suppress pain, such as (lidocaine, morphine, etc.), but the effect is short-lived, poor analgesia, and there are certain dependence and side effects. Therefore, the investigation of the treatment of NPP has become an urgent problem in medical, attracting a lot of research attention. P2X7 is dependent on Adenosine triphosphate (ATP) ion channel receptors and has dual functions for the development of nerve damage and pain. In this review, we explored the link between the P2X7 receptor (P2X7R) and NPP, providing insight into the P2X7R and NPP, discussing the pathological mechanism of P2 X7R in NPP and the biological characteristics of P2X7R antagonist inhibiting its over-expression for the targeted therapy of NPP.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- The Second Affiliate Hospital. Nanchang University, Nanchang City. Jiangxi Province, China; Basic Medical School, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Zheng-Ming Zhu
- The Second Affiliate Hospital. Nanchang University, Nanchang City. Jiangxi Province, China.
| | - Zeng-Xu Liu
- Basic Medical School, Nanchang University, Nanchang City, Jiangxi Province, China
| |
Collapse
|
68
|
Illes P, Verkhratsky A, Tang Y. Pathological ATPergic Signaling in Major Depression and Bipolar Disorder. Front Mol Neurosci 2020; 12:331. [PMID: 32076399 PMCID: PMC7006450 DOI: 10.3389/fnmol.2019.00331] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/26/2019] [Indexed: 12/11/2022] Open
Abstract
The mood disorders, major depression (MD) and bipolar disorder (BD), have a high lifetime prevalence in the human population and accordingly generate huge costs for health care. Efficient, rapidly acting, and side-effect-free pharmaceuticals are hitherto not available, and therefore, the identification of new therapeutic targets is an imperative task for (pre)clinical research. Such a target may be the purinergic P2X7 receptor (P2X7R), which is localized in the central nervous system (CNS) at microglial and neuroglial cells mediating neuroinflammation. MD and BD are due to neuroinflammation caused in the first line by the release of the pro-inflammatory cytokine interleukin-1β (IL-1β) from the microglia. IL-1β in turn induces the secretion of corticotropin-releasing hormone (CRH) and in consequence the secretion of adrenocorticotropic hormone (ACTH) and cortisol, which together with a plethora of further cytokines/chemokines lead to mood disorders. A number of biochemical/molecular biological measurements including the use of P2X7R- or IL-1β-deficient mice confirmed this chain of events. More recent studies showed that a decrease in the astrocytic release of ATP in the prefrontal cortex and hippocampus is a major cause of mood disorders. It is an attractive hypothesis that compensatory increases in P2X7Rs in these areas of the brain are the immediate actuators of MD and BD. Hence, blood-brain barrier-permeable P2X7R antagonists may be promising therapeutic tools to improve depressive disorders in humans.
Collapse
Affiliation(s)
- Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany.,Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom.,Achucarro Centre for Neuroscience, Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
69
|
Insulin-like growth factor-1 inhibits spreading depression-induced trigeminal calcitonin gene related peptide, oxidative stress & neuronal activation in rat. Brain Res 2020; 1732:146673. [PMID: 31978377 DOI: 10.1016/j.brainres.2020.146673] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 11/21/2022]
Abstract
Migraineurs can show brain hyperexcitability and oxidative stress that may promote headache. Since hyperexcitability can enhance oxidative stress which promotes hyperexcitability, ending this feed-back loop may reduce migraine. Neocortical spreading depression, an animal model of migraine begins with hyperexcitability and triggers oxidative stress in the neocortical area involved and in the trigeminal system, which is important to pain pathway nociceptive activation in migraine. Additionally, oxidative stress causes increased trigeminal ganglion calcitonin gene-related peptide release and oxidative stress can reduce spreading depression threshold. Insulin-like growth factor-1 significantly protects against spreading depression in vitro by reducing oxidative stress and it is effective against spreading depression after intranasal delivery to animals. Here, we used adult male rats and extend this work to study the trigeminal system where insulin-like growth factor-1 receptors are highly expressed. Recurrent neocortical spreading depression significantly increased surrogate markers of trigeminal activation - immunostaining for trigeminal ganglion oxidative stress, calcitonin gene related peptide levels and c-fos in the trigeminocervical complex versus sham. These effects were significantly reduced by intranasal delivery of insulin-like growth factor-1 a day before recurrent neocortical spreading depression. Furthermore, intranasal treatment with insulin-like growth factor-1 significantly reduced naïve levels of trigeminal ganglion calcitonin gene related peptide versus sham with no impact on blood glucose levels. Intranasal delivery of insulin-like growth factor-1 not only mitigates neocortical spreading depression, a cause of migraine hyperexcitability modeled in animals, but also when neocortical spreading depression is triggered by supra-threshold stimuli, insulin-like growth factor-1 effectively reduces nociceptive activation in the trigeminal system.
Collapse
|
70
|
Pacheco JM, Hines-Lanham A, Stratton C, Mehos CJ, McCurdy KE, Pinkowski NJ, Zhang H, Shuttleworth CW, Morton RA. Spreading Depolarizations Occur in Mild Traumatic Brain Injuries and Are Associated with Postinjury Behavior. eNeuro 2019; 6:ENEURO.0070-19.2019. [PMID: 31748237 PMCID: PMC6893232 DOI: 10.1523/eneuro.0070-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 01/20/2023] Open
Abstract
Millions of people suffer mild traumatic brain injuries (mTBIs) every year, and there is growing evidence that repeated injuries can result in long-term pathology. The acute symptoms of these injuries may or may not include the loss of consciousness but do include disorientation, confusion, and/or the inability to concentrate. Most of these acute symptoms spontaneously resolve within a few hours or days. However, the underlying physiological and cellular mechanisms remain unclear. Spreading depolarizations (SDs) are known to occur in rodents and humans following moderate and severe TBIs, and SDs have long been hypothesized to occur in more mild injuries. Using a closed skull impact model, we investigated the presence of SDs immediately following a mTBI. Animals remained motionless for multiple minutes following an impact and once recovered had fewer episodes of movement. We recorded the defining electrophysiological properties of SDs, including the large extracellular field potential shifts and suppression of high-frequency cortical activity. Impact-induced SDs were also associated with a propagating wave of reduced cerebral blood flow (CBF). In the wake of the SD, there was a prolonged period of reduced CBF that recovered in approximately 90 min. Similar to SDs in more severe injuries, the impact-induced SDs could be blocked with ketamine. Interestingly, impacts at a slower velocity did not produce the prolonged immobility and did not initiate SDs. Our data suggest that SDs play a significant role in mTBIs and SDs may contribute to the acute symptoms of mTBIs.
Collapse
Affiliation(s)
- Johann M Pacheco
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Ashlyn Hines-Lanham
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Claire Stratton
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Carissa J Mehos
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Kathryn E McCurdy
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Natalie J Pinkowski
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Haikun Zhang
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Russell A Morton
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131
| |
Collapse
|
71
|
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: 8.8] [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
|
72
|
Abstract
PURPOSE OF REVIEW This article discusses the basic mechanisms of migraine aura and its clinical significance based upon evidence from human studies and animal models. RECENT FINDINGS Prospective clinical studies have reinforced the understanding that migraine aura is highly variable from one individual to the next as well as from attack to attack in an individual. While migraine with aura clearly has a higher heritability than migraine without aura, population studies have not identified specific genes that underlie this heritability for typical migraine with aura. Imaging studies reveal hypoperfusion associated with migraine aura, although the timing and distribution of this hypoperfusion is not strictly correlated with migraine symptoms. Mapping of migraine visual aura symptoms onto the visual cortex suggests that the mechanisms underlying the aura propagate in a linear fashion along gyri or sulci rather than as a concentric wave and also suggests that aura may propagate in the absence of clinical symptoms. Cortical spreading depression in animal models continues to be a translational model for migraine, and the study of spreading depolarizations in the injured human brain has provided new insight into potential mechanisms of cortical spreading depression in migraine. Migraine with aura has multiple comorbidities including patent foramen ovale, stroke, and psychiatric disorders; the shared mechanisms underlying these comorbidities remains a topic of active investigation. SUMMARY Although it occurs in the minority of patients with migraine, aura may have much to teach us about basic mechanisms of migraine. In addition, its occurrence may influence clinical management regarding comorbid conditions and acute and preventive therapy.
Collapse
|
73
|
The metabolic face of migraine - from pathophysiology to treatment. Nat Rev Neurol 2019; 15:627-643. [PMID: 31586135 DOI: 10.1038/s41582-019-0255-4] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
Migraine can be regarded as a conserved, adaptive response that occurs in genetically predisposed individuals with a mismatch between the brain's energy reserve and workload. Given the high prevalence of migraine, genotypes associated with the condition seem likely to have conferred an evolutionary advantage. Technological advances have enabled the examination of different aspects of cerebral metabolism in patients with migraine, and complementary animal research has highlighted possible metabolic mechanisms in migraine pathophysiology. An increasing amount of evidence - much of it clinical - suggests that migraine is a response to cerebral energy deficiency or oxidative stress levels that exceed antioxidant capacity and that the attack itself helps to restore brain energy homeostasis and reduces harmful oxidative stress levels. Greater understanding of metabolism in migraine offers novel therapeutic opportunities. In this Review, we describe the evidence for abnormalities in energy metabolism and mitochondrial function in migraine, with a focus on clinical data (including neuroimaging, biochemical, genetic and therapeutic studies), and consider the relationship of these abnormalities with the abnormal sensory processing and cerebral hyper-responsivity observed in migraine. We discuss experimental data to consider potential mechanisms by which metabolic abnormalities could generate attacks. Finally, we highlight potential treatments that target cerebral metabolism, such as nutraceuticals, ketone bodies and dietary interventions.
Collapse
|
74
|
Chen S, Eikermann‐Haerter K. How Imaging Can Help Us Better Understand the Migraine‐Stroke Connection. Headache 2019; 60:217-228. [DOI: 10.1111/head.13664] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Shih‐Pin Chen
- Division of Translational Research Department of Medical Research Taipei Veterans General Hospital Taipei Taiwan
- Department of Neurology Neurological InstituteTaipei Veterans General Hospital Taipei Taiwan
- Institute of Clinical Medicine National Yang‐Ming University School of Medicine Taipei Taiwan
- Brain Research Center National Yang‐Ming University School of Medicine Taipei Taiwan
| | | |
Collapse
|
75
|
Bolay H, Vuralli D, Goadsby PJ. Aura and Head pain: relationship and gaps in the translational models. J Headache Pain 2019; 20:94. [PMID: 31481015 PMCID: PMC6734357 DOI: 10.1186/s10194-019-1042-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/14/2019] [Indexed: 12/13/2022] Open
Abstract
Migraine is a complex brain disorder and initiating events for acute attacks still remain unclear. It seems difficult to explain the development of migraine headache with one mechanism and/or a single anatomical location. Cortical spreading depression (CSD) is recognized as the biological substrate of migraine aura and experimental animal studies have provided mechanisms that possibly link CSD to the activation of trigeminal neurons mediating lateralized head pain. However, some CSD features do not match the clinical features of migraine headache and there are gaps in translating CSD to migraine with aura. Clinical features of migraine headache and results from research are critically evaluated; and consistent and inconsistent findings are discussed according to the known basic features of canonical CSD: typical SD limited to the cerebral cortex as it was originally defined. Alternatively, arguments related to the emergence of SD in other brain structures in addition to the cerebral cortex or CSD initiated dysfunction in the thalamocortical network are proposed. Accordingly, including thalamus, particularly reticular nucleus and higher order thalamic nuclei, which functions as a hub connecting the visual, somatosensory, language and motor cortical areas and subjects to modulation by brain stem projections into the CSD theory, would greatly improve our current understanding of migraine.
Collapse
Affiliation(s)
- Hayrunnisa Bolay
- Department of Neurology and Algology, Gazi University Faculty of Medicine, Besevler, 06510 Ankara, Turkey
- Neuropsychiatry Center, Gazi University, Besevler, Ankara, Turkey
| | - Doga Vuralli
- Neuropsychiatry Center, Gazi University, Besevler, Ankara, Turkey
- Department of Algology, Bakirkoy Sadi Konuk Training and Research Hospital, Bakirkoy, Istanbul, Turkey
| | - Peter J. Goadsby
- Headache Group, Department of Basic and Clinical Neuroscience, King’s College London, London, UK
- NIHR-Wellcome Trust King’s Clinical Research Facility, King’s College Hospital, London, UK
| |
Collapse
|
76
|
Driessen AK. Vagal Afferent Processing by the Paratrigeminal Nucleus. Front Physiol 2019; 10:1110. [PMID: 31555145 PMCID: PMC6722180 DOI: 10.3389/fphys.2019.01110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/12/2019] [Indexed: 12/26/2022] Open
Abstract
The paratrigeminal nucleus is an obscure region in the dorsal lateral medulla, which has been best characterized as a collection of interstitial cells located in the dorsal tip of the spinal trigeminal tract. The paratrigeminal nucleus receives afferent input from the vagus, trigeminal, spinal, and glossopharyngeal nerves, which contribute to its long-known roles in the baroreceptor reflex and nociceptive processing. More recently, studies have shown that this region is also involved in the processing of airway-derived sensory information. Notably, these studies highlight an underappreciated complexity in the neuronal content and circuit connectivity of the paratrigeminal nucleus. However, much remains to be understood about how paratrigeminal processing of vagal afferents is altered in disease. The aim of the present review is to provide an update of the current understanding of vagal afferent processing in the paratrigeminal nucleus and to explore how dysregulation at this site may contribute to vagal sensory neural dysfunction during disease.
Collapse
Affiliation(s)
- Alexandria K Driessen
- School of Biomedical Science, Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
77
|
Yemisci M, Eikermann-Haerter K. Aura and Stroke: relationship and what we have learnt from preclinical models. J Headache Pain 2019; 20:63. [PMID: 31142262 PMCID: PMC6734247 DOI: 10.1186/s10194-019-1016-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/19/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Population-based studies have highlighted a close relationship between migraine and stroke. Migraine, especially with aura, is a risk factor for both ischemic and hemorrhagic stroke. Interestingly, stroke risk is highest for migraineurs who are young and otherwise healthy. MAIN BODY Preclinical models have provided us with possible mechanisms to explain the increased vulnerability of migraineurs' brains towards ischemia and suggest a key role for enhanced cerebral excitability and increased incidence of microembolic events. Spreading depolarization (SD), a slowly propagating wave of neuronal depolarization, is the electrophysiologic event underlying migraine aura and a known headache trigger. Increased SD susceptibility has been demonstrated in migraine animal models, including transgenic mice carrying human mutations for the migraine-associated syndrome CADASIL and familial hemiplegic migraine (type 1 and 2). Upon experimentally induced SD, these mice develop aura-like neurological symptoms, akin to patients with the respective mutations. Migraine mutant mice also exhibit an increased frequency of ischemia-triggered SDs upon experimental stroke, associated with accelerated infarct growth and worse outcomes. The severe stroke phenotype can be explained by SD-related downstream events that exacerbate the metabolic mismatch, including pericyte contraction and neuroglial inflammation. Pharmacological suppression of the genetically enhanced SD susceptibility normalizes the stroke phenotype in familial hemiplegic migraine mutant mice. Recent epidemiologic and imaging studies suggest that these preclinical findings can be extrapolated to migraine patients. Migraine patients are at risk for particularly cardioembolic stroke. At the same time, studies suggest an increased incidence of coagulopathy, atrial fibrillation and patent foramen ovale among migraineurs, providing a possible path for microembolic induction of SD and, in rare instances, stroke in hyperexcitable brains. Indeed, recent imaging studies document an accelerated infarct progression with only little potentially salvageable brain tissue in acute stroke patients with a migraine history, suggesting an increased vulnerability towards cerebral ischemia. CONCLUSION Preclinical models suggest a key role for enhanced SD susceptibility and microembolization to explain both the occurrence of migraine attacks and the increased stroke risk in migraineurs. Therapeutic targeting of SD and microembolic events, or potential causes thereof, will be promising for treatment of aura and may also prevent ischemic infarction in vulnerable brains.
Collapse
Affiliation(s)
- Muge Yemisci
- Institute of Neurological Sciences and Psychiatry, and Faculty of Medicine, Department of Neurology, Hacettepe University, Ankara, Turkey
| | | |
Collapse
|
78
|
Harriott AM, Takizawa T, Chung DY, Chen SP. Spreading depression as a preclinical model of migraine. J Headache Pain 2019; 20:45. [PMID: 31046659 PMCID: PMC6734429 DOI: 10.1186/s10194-019-1001-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/18/2019] [Indexed: 01/12/2023] Open
Abstract
Spreading depression (SD) is a slowly propagating wave of near-complete depolarization of neurons and glial cells across the cortex. SD is thought to contribute to the underlying pathophysiology of migraine aura, and possibly also an intrinsic brain activity causing migraine headache. Experimental models of SD have recapitulated multiple migraine-related phenomena and are considered highly translational. In this review, we summarize conventional and novel methods to trigger SD, with specific focus on optogenetic methods. We outline physiological triggers that might affect SD susceptibility, review a multitude of physiological, biochemical, and behavioral consequences of SD, and elaborate their relevance to migraine pathophysiology. The possibility of constructing a recurrent episodic or chronic migraine model using SD is also discussed.
Collapse
Affiliation(s)
- Andrea M Harriott
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Tsubasa Takizawa
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - David Y Chung
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Shih-Pin Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan. .,Brain Research Center, National Yang-Ming University, Taipei, Taiwan. .,Division of Translational Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan. .,Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
79
|
Szabó Í, M. Tóth O, Török Z, Varga DP, Menyhárt Á, Frank R, Hantosi D, Hunya Á, Bari F, Horváth I, Vigh L, Farkas E. The impact of dihydropyridine derivatives on the cerebral blood flow response to somatosensory stimulation and spreading depolarization. Br J Pharmacol 2019; 176:1222-1234. [PMID: 30737967 PMCID: PMC6468258 DOI: 10.1111/bph.14611] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/07/2019] [Accepted: 01/11/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE A new class of dihydropyridine derivatives, which act as co-inducers of heat shock protein but are devoid of calcium channel antagonist and vasodilator effects, has recently been developed with the purpose of selectively targeting neurodegeneration. Here, we evaluated the action of one of these novel compounds LA1011 on neurovascular coupling in the ischaemic rat cerebral cortex. As a reference, we applied nimodipine, a vasodilator dihydropyridine and well-known calcium channel antagonist. EXPERIMENTAL APPROACH Rats were treated with LA1011 or nimodipine, either by chronic, systemic (LA1011), or acute, local administration (LA1011 and nimodipine). In the latter treatment group, global forebrain ischaemia was induced in half of the animals by bilateral common carotid artery occlusion under isoflurane anaesthesia. Functional hyperaemia in the somatosensory cortex was created by mechanical stimulation of the contralateral whisker pad under α-chloralose anaesthesia. Spreading depolarization (SD) events were elicited subsequently by 1 M KCl. Local field potential and cerebral blood flow (CBF) in the parietal somatosensory cortex were monitored by electrophysiology and laser Doppler flowmetry. KEY RESULTS LA1011 did not alter CBF, but intensified SD, presumably indicating the co-induction of heat shock proteins, and, perhaps an anti-inflammatory effect. Nimodipine attenuated evoked potentials and SD. In addition to the elevation of baseline CBF, nimodipine augmented hyperaemia in response to both somatosensory stimulation and SD, particularly under ischaemia. CONCLUSIONS AND IMPLICATIONS In contrast to the CBF improvement achieved with nimodipine, LA1011 seems not to have discernible cerebrovascular effects but may up-regulate the stress response.
Collapse
Affiliation(s)
- Írisz Szabó
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and InformaticsUniversity of SzegedSzegedHungary
| | - Orsolya M. Tóth
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and InformaticsUniversity of SzegedSzegedHungary
| | - Zsolt Török
- Institute of Biochemistry, Biological Research CentreHungarian Academy of SciencesSzegedHungary
- LipidArt Research and Development Ltd.SzegedHungary
| | - Dániel Péter Varga
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and InformaticsUniversity of SzegedSzegedHungary
| | - Ákos Menyhárt
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and InformaticsUniversity of SzegedSzegedHungary
| | - Rita Frank
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and InformaticsUniversity of SzegedSzegedHungary
| | - Dóra Hantosi
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and InformaticsUniversity of SzegedSzegedHungary
| | - Ákos Hunya
- LipidArt Research and Development Ltd.SzegedHungary
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and InformaticsUniversity of SzegedSzegedHungary
| | - Ibolya Horváth
- Institute of Biochemistry, Biological Research CentreHungarian Academy of SciencesSzegedHungary
| | - László Vigh
- Institute of Biochemistry, Biological Research CentreHungarian Academy of SciencesSzegedHungary
| | - Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and InformaticsUniversity of SzegedSzegedHungary
| |
Collapse
|
80
|
Yalcin N, Chen SP, Yu ES, Liu TT, Yen JC, Atalay YB, Qin T, Celik F, van den Maagdenberg AM, Moskowitz MA, Ayata C, Eikermann-Haerter K. Caffeine does not affect susceptibility to cortical spreading depolarization in mice. J Cereb Blood Flow Metab 2019; 39:740-750. [PMID: 29651899 PMCID: PMC6446422 DOI: 10.1177/0271678x18768955] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Several factors that modulate migraine, a common primary headache disorder, also affect susceptibility to cortical spreading depolarization (CSD). CSD is a wave of neuronal and glial depolarization and thought to underlie the migraine aura and possibly headache. Here, we tested whether caffeine, known to alleviate or trigger headache after acute exposure or chronic use/withdrawal, respectively, modulates CSD. We injected C57BL/6J mice with caffeine (30, 60, or 120 mg/kg; i.p.) once ( acute) or twice per day for one or two weeks ( chronic). Susceptibility to CSD was evaluated by measuring the electrical CSD threshold and by assessing KCl-induced CSD. Simultaneous laser Doppler flowmetry was used to assess CSD-induced cortical blood flow changes. Recordings were performed 15 min after caffeine/vehicle administration, or 24 h after the last dose of chronic caffeine in the withdrawal group. The latter paradigm was also tested in mice carrying the familial hemiplegic migraine type 1 R192Q missense mutation, considered a valid migraine model. Neither acute/chronic administration nor withdrawal of caffeine affected CSD susceptibility or related cortical blood flow changes, either in WT or R192Q mice. Hence, adverse or beneficial effects of caffeine on headache seem unrelated to CSD pathophysiology, consistent with the non-migrainous clinical presentation of caffeine-related headache.
Collapse
Affiliation(s)
- Nilufer Yalcin
- 1 Neurovascular Research Laboratory, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Shih-Pin Chen
- 2 Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,3 Department of Medical Research, Division of Translational Research, Taipei Veterans General Hospital, Taipei, Taiwan.,4 Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Esther S Yu
- 1 Neurovascular Research Laboratory, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Tzu-Ting Liu
- 5 Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Jiin-Cherng Yen
- 5 Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Yahya B Atalay
- 1 Neurovascular Research Laboratory, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Tao Qin
- 1 Neurovascular Research Laboratory, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Furkan Celik
- 1 Neurovascular Research Laboratory, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Arn Mjm van den Maagdenberg
- 6 Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.,7 Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael A Moskowitz
- 1 Neurovascular Research Laboratory, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Cenk Ayata
- 1 Neurovascular Research Laboratory, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA.,8 Department of Neurology, Harvard Medical School, Stroke Service and Neuroscience Intensive Care Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Katharina Eikermann-Haerter
- 1 Neurovascular Research Laboratory, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
81
|
Illes P, Rubini P, Huang L, Tang Y. The P2X7 receptor: a new therapeutic target in Alzheimer’s disease. Expert Opin Ther Targets 2019; 23:165-176. [DOI: 10.1080/14728222.2019.1575811] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Patrizia Rubini
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Lumei Huang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of TCM, Chengdu, China
| |
Collapse
|
82
|
Nurkhametova D, Kudryavtsev I, Guselnikova V, Serebryakova M, Giniatullina RR, Wojciechowski S, Tore F, Rizvanov A, Koistinaho J, Malm T, Giniatullin R. Activation of P2X7 Receptors in Peritoneal and Meningeal Mast Cells Detected by Uptake of Organic Dyes: Possible Purinergic Triggers of Neuroinflammation in Meninges. Front Cell Neurosci 2019; 13:45. [PMID: 30814932 PMCID: PMC6381076 DOI: 10.3389/fncel.2019.00045] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/28/2019] [Indexed: 12/24/2022] Open
Abstract
Extracellular ATP activates inflammasome and triggers the release of multiple cytokines in various immune cells, a process primarily mediated by P2X7 receptors. However, the expression and functional properties of P2X7 receptors in native mast cells in tissues such as meninges where migraine pain originates from have not been explored. Here we report a novel model of murine cultured meningeal mast cells and using these, as well as easily accessible peritoneal mast cells, studied the mechanisms of ATP-mediated mast cell activation. We show that ATP induced a time and dose-dependent activation of peritoneal mast cells as analyzed by the uptake of organic dye YO-PRO1 as well as 4,6-diamidino-2-phenylindole (DAPI). Both YO-PRO1 and DAPI uptake in mast cells was mediated by the P2X7 subtype of ATP receptors as demonstrated by the inhibitory effect of P2X7 antagonist A839977. Consistent with this, significant YO-PRO1 uptake was promoted by the P2X7 agonist 2′,3′-O-(benzoyl-4-benzoyl)-ATP (BzATP). Extracellular ATP-induced degranulation of native and cultured meningeal mast cells was shown with Toluidine Blue staining. Taken together, these data demonstrate the important contribution of P2X7 receptors to ATP-driven activation of mast cells, suggesting these purinergic mechanisms as potential triggers of neuroinflammation and pain sensitization in migraine.
Collapse
Affiliation(s)
- Dilyara Nurkhametova
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia.,A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Igor Kudryavtsev
- Department of Immunology, Institute of Experimental Medicine, St. Petersburg, Russia.,Department of Fundamental Medicine, Far Eastern Federal University, Vladivostok, Russia
| | - Valeriia Guselnikova
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Department of General and Special Morphology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Maria Serebryakova
- Department of Immunology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Raisa R Giniatullina
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sara Wojciechowski
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Fatma Tore
- School of Medicine, Biruni University, Istanbul, Turkey
| | - Albert Rizvanov
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Jari Koistinaho
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tarja Malm
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Rashid Giniatullin
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia.,A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
83
|
Kilic K, Karatas H, Dönmez-Demir B, Eren-Kocak E, Gursoy-Ozdemir Y, Can A, Petit JM, Magistretti PJ, Dalkara T. Inadequate brain glycogen or sleep increases spreading depression susceptibility. Ann Neurol 2019; 83:61-73. [PMID: 29244233 DOI: 10.1002/ana.25122] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/23/2017] [Accepted: 12/12/2017] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Glycogen in astrocyte processes contributes to maintenance of low extracellular glutamate and K+ concentrations around excitatory synapses. Sleep deprivation (SD), a common migraine trigger, induces transcriptional changes in astrocytes, reducing glycogen breakdown. We hypothesize that when glycogen utilization cannot match synaptic energy demand, extracellular K+ can rise to levels that activate neuronal pannexin-1 channels and downstream inflammatory pathway, which might be one of the mechanisms initiating migraine headaches. METHODS We suppressed glycogen breakdown by inhibiting glycogen phosphorylation with 1,4-dideoxy-1,4-imino-D-arabinitol (DAB) and by SD. RESULTS DAB caused neuronal pannexin-1 large pore opening and activation of the downstream inflammatory pathway as shown by procaspase-1 cleavage and HMGB1 release from neurons. Six-hour SD induced pannexin-1 mRNA. DAB and SD also lowered the cortical spreading depression (CSD) induction threshold, which was reversed by glucose or lactate supplement, suggesting that glycogen-derived energy substrates are needed to prevent CSD generation. Supporting this, knocking down the neuronal lactate transporter MCT2 with an antisense oligonucleotide or inhibiting glucose transport from vessels to astrocytes with intracerebroventricularly delivered phloretin reduced the CSD threshold. In vivo recordings with a K+ -sensitive/selective fluoroprobe, Asante Potassium Green-4, revealed that DAB treatment or SD caused a significant rise in extracellular K+ during whisker stimulation, illustrating the critical role of glycogen in extracellular K+ clearance. INTERPRETATION Synaptic metabolic stress caused by insufficient glycogen-derived energy substrate supply can activate neuronal pannexin-1 channels as well as lower the CSD threshold. Therefore, conditions that limit energy supply to synapses (eg, SD) may predispose to migraine attacks, as suggested by genetic studies associating glucose or lactate transporter deficiency with migraine. Ann Neurol 2018;83:61-73.
Collapse
Affiliation(s)
- Kivilcim Kilic
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Buket Dönmez-Demir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Emine Eren-Kocak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Yasemin Gursoy-Ozdemir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Current address for Dr Gursoy-Ozdemir: Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
| | - Alp Can
- Department of Histology and Embryology, School of Medicine, Ankara University, Ankara, Turkey
| | - Jean-Marie Petit
- Center for Psychiatric Neuroscience, Department of Psychiatry, University Hospital of the Canton of Vaud (CHUV), Prilly, Switzerland
| | - Pierre J Magistretti
- King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.,Brain Mind Institute, Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Turgay Dalkara
- Department of Neurology, Faculty of Medicine and Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
84
|
Susceptibility of the cerebral cortex to spreading depolarization in neurological disease states: The impact of aging. Neurochem Int 2018; 127:125-136. [PMID: 30336178 DOI: 10.1016/j.neuint.2018.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/10/2018] [Accepted: 10/13/2018] [Indexed: 12/17/2022]
Abstract
Secondary injury following acute brain insults significantly contributes to poorer neurological outcome. The spontaneous, recurrent occurrence of spreading depolarization events (SD) has been recognized as a potent secondary injury mechanism in subarachnoid hemorrhage, malignant ischemic stroke and traumatic brain injury. In addition, SD is the underlying mechanism of the aura symptoms of migraineurs. The susceptibility of the nervous tissue to SD is subject to the metabolic status of the tissue, the ionic composition of the extracellular space, and the functional status of ion pumps, voltage-gated and other cation channels, glutamate receptors and excitatory amino acid transporters. All these mechanisms tune the excitability of the nervous tissue. Aging has also been found to alter SD susceptibility, which appears to be highest at young adulthood, and decline over the aging process. The lower susceptibility of the cerebral gray matter to SD in the old brain may be caused by the age-related impairment of mechanisms implicated in ion translocations between the intra- and extracellular compartments, glutamate signaling and surplus potassium and glutamate clearance. Even though the aging nervous tissue is thus less able to sustain SD, the consequences of SD recurrence in the old brain have proven to be graver, possibly leading to accelerated lesion maturation. Taken that recurrent SDs may pose an increased burden in the aging injured brain, the benefit of therapeutic approaches to restrict SD generation and propagation may be particularly relevant for elderly patients.
Collapse
|
85
|
Abstract
The connexin family of channel-forming proteins is present in every tissue type in the human anatomy. Connexins are best known for forming clustered intercellular channels, structurally known as gap junctions, where they serve to exchange members of the metabolome between adjacent cells. In their single-membrane hemichannel form, connexins can act as conduits for the passage of small molecules in autocrine and paracrine signalling. Here, we review the roles of connexins in health and disease, focusing on the potential of connexins as therapeutic targets in acquired and inherited diseases as well as wound repair, while highlighting the associated clinical challenges.
Collapse
|
86
|
Nurkhametova D, Kudryavtsev I, Khayrutdinova O, Serebryakova M, Altunbaev R, Malm T, Giniatullin R. Purinergic Profiling of Regulatory T-cells in Patients With Episodic Migraine. Front Cell Neurosci 2018; 12:326. [PMID: 30319363 PMCID: PMC6167492 DOI: 10.3389/fncel.2018.00326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/07/2018] [Indexed: 01/03/2023] Open
Abstract
Objectives: Immune responses in migraine are poorly characterized, yet implicated in the disease pathogenesis. This study was carried out to characterize purinergic profiles of T-cells in patients with episodic migraine without aura (MWoA) to provide mechanistic evidence for ATP and adenosine involvement in modulation of immune regulation in migraine. Methods: Peripheral blood samples were obtained from patients with migraine (n = 16) and age-matched control subjects (n = 21). Subsets of T-cells were identified by flow cytometry based on specific membrane markers. Results: Migraine patients showed reduced total T-cell counts in the peripheral blood. Whereas the total number of CD3+CD4+, CD3+CD8+, or regulatory T lymphocytes (Treg) was not changed, the proportion of Treg CD45R0+CD62L- and CD45R0-CD62L- cells was increased. Interestingly, in migraine, less Treg cells expressed CD39 and CD73 suggesting disrupted ATP breakdown to adenosine. The negative correlations were observed between the duration of migraine and the relative number of CD73+CD39- Tregs and total number of CD73-positive CD45R0+CD62L+ Tregs. Conclusion: Obtained data indicate that T-cell populations are altered in episodic migraine and suggest the involvement of Tregs in the pathophysiology of this disorder. Reduced expression of CD39 and CD73 suggests promotion of ATP-dependent pro-inflammatory and reduction of adenosine-mediated anti-inflammatory mechanisms in migraine.
Collapse
Affiliation(s)
- Dilyara Nurkhametova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | - Igor Kudryavtsev
- Department of Immunology, Institute of Experimental Medicine, St. Petersburg, Russia
- Department of Fundamental Medicine, Far Eastern Federal University, Vladivostok, Russia
| | - Olga Khayrutdinova
- Department of Neurology and Rehabilitation, Kazan State Medical University, Kazan, Russia
| | - Maria Serebryakova
- Department of Immunology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Rashid Altunbaev
- Department of Neurology and Rehabilitation, Kazan State Medical University, Kazan, Russia
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
87
|
|
88
|
Good ME, Eucker SA, Li J, Bacon HM, Lang SM, Butcher JT, Johnson TJ, Gaykema RP, Patel MK, Zuo Z, Isakson BE. Endothelial cell Pannexin1 modulates severity of ischemic stroke by regulating cerebral inflammation and myogenic tone. JCI Insight 2018; 3:96272. [PMID: 29563335 PMCID: PMC5926909 DOI: 10.1172/jci.insight.96272] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 02/16/2018] [Indexed: 12/24/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality in the US; however, there currently exists only one effective acute pharmacological therapeutic intervention. Purinergic signaling has been shown to regulate vascular function and pathological processes, including inflammation and arterial myogenic reactivity, and plays a role in ischemic stroke outcome. Purinergic signaling requires extracellular purines; however, the mechanism of purine release from cells is unclear. Pannexin1 (Panx1) channels are potentially novel purine release channels expressed throughout the vascular tree that couples regulated purine release with purinergic signaling. Therefore, we examined the role of smooth muscle and endothelial cell Panx1, using conditional cell type-specific transgenic mice, in cerebral ischemia/reperfusion injury outcomes. Deletion of endothelial cell Panx1, but not smooth muscle cell Panx1, significantly reduced cerebral infarct volume after ischemia/reperfusion. Infiltration of leukocytes into brain tissue and development of cerebral myogenic tone were both significantly reduced when mice lacked endothelial Panx1. Panx1 regulation of myogenic tone was unique to the cerebral circulation, as mesenteric myogenic reactivity and blood pressure were independent of endothelial Panx1. Overall, deletion of endothelial Panx1 mitigated cerebral ischemic injury by reducing inflammation and myogenic tone development, indicating that endothelial Panx1 is a possible novel target for therapeutic intervention of ischemic stroke.
Collapse
Affiliation(s)
- Miranda E Good
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Stephanie A. Eucker
- Division of Emergency Medicine, Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Jun Li
- Department of Anesthesiology and
| | - Hannah M. Bacon
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Susan M. Lang
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Joshua T. Butcher
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Tyler J. Johnson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | | | | | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
89
|
Bu F, Wang Y, Jiang L, Ma D, Quinn JP, Wang M. Sarcoma family kinase activity is required for cortical spreading depression. Cephalalgia 2017; 38:1748-1758. [PMID: 29239212 DOI: 10.1177/0333102417748572] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objectives Sarcoma family kinase activity is associated with multiple diseases including ischemia and cancer; however, its role in the mechanism of migraine aura has been less well characterised. This study aims to investigate whether sarcoma family kinase is required for cortical spreading depression. Methods Cortical spreading depression was induced by topical application of K+ to the cerebral cortex and was monitored using electrophysiology in rats, and intrinsic optical signal in mouse brain slices. Drugs were perfused into the contralateral cerebral ventricle for pharmacological manipulations in rats. Western blot analysis was used for detecting the level of phosphorylated, and total, sarcoma family kinase in the ipsilateral cortex of rats. Key results The data demonstrate that a single cortical spreading depression in rats induced ipsilateral cortical sarcoma family kinase phosphorylation at the Y416 site. Deactivation of sarcoma family kinase by its inhibitor (3-(4-chlorophenyl) 1-(1,1-dimethylethyl)-1 H-pyrazolo[3,4- dpyrimidin-4-amine) suppressed the elevated enzyme activity and cortical susceptibility to cortical spreading depression. Interestingly, the inhibitory effect of the N-methyl-D-aspartate receptor antagonist NVP-AAM077 on cortical spreading depression was reversed by the sarcoma family kinase activator pYEEI (EPQY(PO3H2)EEEIPIYL), suggesting a link between this enzyme and N-methyl-D-aspartate receptors. Similarly, after deactivation of sarcoma family kinase, a reduction of sarcoma family kinase phosphorylation and cortical susceptibility to cortical spreading depression was observed with NVP-AAM077. Conclusions We conclude that activation of sarcoma family kinase is required for cortical spreading depression, and this process is regulated by recruiting N-methyl-D-aspartate receptors. This study provides novel insight for sarcoma family kinase function in the mechanism of migraine aura.
Collapse
Affiliation(s)
- Fan Bu
- 1 Department of Biological Sciences, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China.,2 Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China
| | - Yan Wang
- 2 Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China
| | - Liwen Jiang
- 1 Department of Biological Sciences, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China.,2 Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China
| | - Dongqing Ma
- 1 Department of Biological Sciences, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China.,2 Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China
| | - John P Quinn
- 3 Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Minyan Wang
- 1 Department of Biological Sciences, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China.,2 Centre for Neuroscience, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China
| |
Collapse
|
90
|
Unekawa M, Ikeda K, Tomita Y, Kawakami K, Suzuki N. Enhanced susceptibility to cortical spreading depression in two types of Na +,K +-ATPase α2 subunit-deficient mice as a model of familial hemiplegic migraine 2. Cephalalgia 2017; 38:1515-1524. [PMID: 29041816 DOI: 10.1177/0333102417738249] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Patients with familial hemiplegic migraine type 2 (FHM2) have a mutated ATP1A2 gene (encoding Na+,K+-ATPase α2 subunit) and show prolonged migraine aura. Cortical spreading depression (CSD), which involves mass depolarization of neurons and astrocytes that propagates slowly through the gray matter, is profoundly related to aura. Methods In two types of Atp1a2-defective heterozygous mice, Atp1a2tm1Kwk (C-KO) and Atp1a2tm2Kwk (N-KO), the sensitivity and responsiveness to CSD were examined under urethane anesthesia. Results In both cases, heterozygotes exhibited a low threshold for induction of CSD, faster propagation rate, slower recovery from DC deflection, and profound suppression of the electroencephalogram, compared to wild-type mice. A high dose of KCl elicited repeated CSDs for a longer period, with a tendency for a greater frequency of CSD occurrence in heterozygotes. The difference of every endpoint was slightly greater in N-KO than C-KO. Change of regional cerebral blood flow in response to CSD showed no significant difference. Conclusion Heterozygotes of Atp1a2-defective mice simulating FHM2 demonstrated high susceptibility to CSD rather than cortical vasoreactivity, and these effects may differ depending upon the knockout strategy for the gene disruption. These results suggest that patients with FHM2 may exhibit high susceptibility to CSD, resulting in migraine.
Collapse
Affiliation(s)
- Miyuki Unekawa
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Ikeda
- 2 Division of Biology, Hyogo College of Medicine, Nishinomiya, Japan.,3 Division of Biology, Center for Molecular Medicine, Jichi Medical School, Shimotsuke, Japan
| | - Yutaka Tomita
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kiyoshi Kawakami
- 3 Division of Biology, Center for Molecular Medicine, Jichi Medical School, Shimotsuke, Japan
| | - Norihiro Suzuki
- 1 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
91
|
Chen SP, Ayata C. Novel Therapeutic Targets Against Spreading Depression. Headache 2017; 57:1340-1358. [PMID: 28842982 DOI: 10.1111/head.13154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/07/2017] [Accepted: 05/08/2017] [Indexed: 12/11/2022]
Abstract
Migraine is among the most prevalent and disabling neurological diseases in the world. Cortical spreading depression (SD) is an intense wave of neuronal and glial depolarization underlying migraine aura, and a headache trigger, which has been used as an experimental platform for drug screening in migraine. Here, we provide an overview of novel therapeutic targets that show promise to suppress SD, such as acid-sensing ion channels, casein kinase Iδ, P2X7-pannexin 1 complex, and neuromodulation, and outline the experimental models and essential quality measures for rigorous and reproducible efficacy testing.
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.,Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - 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, Charlestown, MA, USA
| |
Collapse
|