1
|
Chichorro JG, Gambeta E, Baggio DF, Zamponi GW. Voltage-gated Calcium Channels as Potential Therapeutic Targets in Migraine. THE JOURNAL OF PAIN 2024; 25:104514. [PMID: 38522594 DOI: 10.1016/j.jpain.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024]
Abstract
Migraine is a complex and highly incapacitating neurological disorder that affects around 15% of the general population with greater incidence in women, often at the most productive age of life. Migraine physiopathology is still not fully understood, but it involves multiple mediators and events in the trigeminovascular system and the central nervous system. The identification of calcitonin gene-related peptide as a key mediator in migraine physiopathology has led to the development of effective and highly selective antimigraine therapies. However, this treatment is neither accessible nor effective for all migraine sufferers. Thus, a better understanding of migraine mechanisms and the identification of potential targets are still clearly warranted. Voltage-gated calcium channels (VGCCs) are widely distributed in the trigeminovascular system, and there is accumulating evidence of their contribution to the mechanisms associated with headache pain. Several drugs used in migraine abortive or prophylactic treatment target VGCCs, which probably contributes to their analgesic effect. This review aims to summarize the current evidence of VGGC contribution to migraine physiopathology and to discuss how current pharmacological options for migraine treatment interfere with VGGC function. PERSPECTIVE: Calcitonin gene-related peptide (CGRP) represents a major migraine mediator, but few studies have investigated the relationship between CGRP and VGCCs. CGRP release is calcium channel-dependent and VGGCs are key players in familial migraine. Further studies are needed to determine whether VGCCs are suitable molecular targets for treating migraine.
Collapse
Affiliation(s)
- Juliana G Chichorro
- Biological Sciences Sector, Department of Pharmacology, Federal University of Parana, Curitiba, Parana, Brazil.
| | - Eder Gambeta
- Cumming School of Medicine, Department of Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Darciane F Baggio
- Biological Sciences Sector, Department of Pharmacology, Federal University of Parana, Curitiba, Parana, Brazil
| | - Gerald W Zamponi
- Cumming School of Medicine, Department of Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
2
|
Loya-Lopez SI, Allen HN, Duran P, Calderon-Rivera A, Gomez K, Kumar U, Shields R, Zeng R, Dwivedi A, Saurabh S, Korczeniewska OA, Khanna R. Intranasal CRMP2-Ubc9 inhibitor regulates Na V 1.7 to alleviate trigeminal neuropathic pain. Pain 2024; 165:573-588. [PMID: 37751532 PMCID: PMC10922202 DOI: 10.1097/j.pain.0000000000003053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 09/28/2023]
Abstract
ABSTRACT Dysregulation of voltage-gated sodium Na V 1.7 channels in sensory neurons contributes to chronic pain conditions, including trigeminal neuropathic pain. We previously reported that chronic pain results in part from increased SUMOylation of collapsin response mediator protein 2 (CRMP2), leading to an increased CRMP2/Na V 1.7 interaction and increased functional activity of Na V 1.7. Targeting this feed-forward regulation, we developed compound 194 , which inhibits CRMP2 SUMOylation mediated by the SUMO-conjugating enzyme Ubc9. We further demonstrated that 194 effectively reduces the functional activity of Na V 1.7 channels in dorsal root ganglia neurons and alleviated inflammatory and neuropathic pain. Here, we used a comprehensive array of approaches, encompassing biochemical, pharmacological, genetic, electrophysiological, and behavioral analyses, to assess the functional implications of Na V 1.7 regulation by CRMP2 in trigeminal ganglia (TG) neurons. We confirmed the expression of Scn9a , Dpysl2 , and UBE2I within TG neurons. Furthermore, we found an interaction between CRMP2 and Na V 1.7, with CRMP2 being SUMOylated in these sensory ganglia. Disrupting CRMP2 SUMOylation with compound 194 uncoupled the CRMP2/Na V 1.7 interaction, impeded Na V 1.7 diffusion on the plasma membrane, and subsequently diminished Na V 1.7 activity. Compound 194 also led to a reduction in TG neuron excitability. Finally, when intranasally administered to rats with chronic constriction injury of the infraorbital nerve, 194 significantly decreased nociceptive behaviors. Collectively, our findings underscore the critical role of CRMP2 in regulating Na V 1.7 within TG neurons, emphasizing the importance of this indirect modulation in trigeminal neuropathic pain.
Collapse
Affiliation(s)
- Santiago I. Loya-Lopez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Heather N. Allen
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Upasana Kumar
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Newark, NJ 07101, United States of America
| | - Rory Shields
- Rutgers School of Graduate Studies, Newark Health Science Campus, Newark, NJ 07101, United States of America
| | - Rui Zeng
- Department of Chemistry, College of Arts and Sciences, New York University, 100 Washington Square East, New York, NY 10003, United States of America
| | - Akshat Dwivedi
- Department of Chemistry, College of Arts and Sciences, New York University, 100 Washington Square East, New York, NY 10003, United States of America
| | - Saumya Saurabh
- Department of Chemistry, College of Arts and Sciences, New York University, 100 Washington Square East, New York, NY 10003, United States of America
| | - Olga A. Korczeniewska
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Newark, NJ 07101, United States of America
- Rutgers School of Graduate Studies, Newark Health Science Campus, Newark, NJ 07101, United States of America
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
- Department of Neuroscience and Physiology and Neuroscience Institute, School of Medicine, New York University, New York, NY, 10010, USA
| |
Collapse
|
3
|
Pearl NZ, Babin CP, Catalano NT, Blake JC, Ahmadzadeh S, Shekoohi S, Kaye AD. Narrative Review of Topiramate: Clinical Uses and Pharmacological Considerations. Adv Ther 2023; 40:3626-3638. [PMID: 37368102 DOI: 10.1007/s12325-023-02586-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023]
Abstract
Due to the diverse mechanisms of action of antiseizure drugs, there has been a rise in prescriptions of these drugs for non-epileptic pathologies. One drug that is now being used for a variety of conditions is topiramate. This is a narrative review that used PubMed, Google Scholar, MEDLINE, and ScienceDirect to review literature on the clinical and pharmacologic properties of topiramate. Topiramate is a commonly prescribed second-generation antiseizure drug. The drug works through multiple pathways to prevent seizures. In this regard, topiramate blocks sodium and calcium voltage-gated channels, inhibits glutamate receptors, enhances gamma-aminobutyric acid (GABA) receptors, and inhibits carbonic anhydrase. Topiramate is approved by the Food and Drug Administration (FDA) for epilepsy treatment and migraine prophylaxis. Topiramate in combination with phentermine is also FDA-approved for weight loss in patients with a body mass index (BMI) > 30. The current target dosing for topiramate monotherapy is 400 mg/day and 100 mg/day to treat epilepsy and migraines, respectively. Commonly reported side effects include paresthesia, confusion, fatigue, dizziness, and change in taste. More uncommon and serious adverse effects can include acute glaucoma, metabolic acidosis, nephrolithiasis, hepatotoxicity, and teratogenicity. Related to a broad side effect profile, physicians prescribing this drug should routinely monitor for side effects and/or toxicity. The present investigation reviews various anti-seizure medications before summarizing indications of topiramate, off-label uses, pharmacodynamics, pharmacokinetics, adverse effects, and drug-drug interactions.
Collapse
Affiliation(s)
- Nathan Z Pearl
- School of Medicine, LSU Health Sciences Center New Orleans, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Caroline P Babin
- School of Medicine, LSU Health Sciences Center New Orleans, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Nicole T Catalano
- School of Medicine, LSU Health Sciences Center New Orleans, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - James C Blake
- School of Medicine, LSU Health Sciences Center New Orleans, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
- Department of Pharmacology, Toxicology, and Neurosciences, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| |
Collapse
|
4
|
Loya-Lopez SI, Allen HN, Duran P, Calderon-Rivera A, Gomez K, Kumar U, Shields R, Zeng R, Dwivedi A, Saurabh S, Korczeniewska OA, Khanna R. Intranasal CRMP2-Ubc9 Inhibitor Regulates Na V 1.7 to Alleviate Trigeminal Neuropathic Pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549195. [PMID: 37502910 PMCID: PMC10370107 DOI: 10.1101/2023.07.16.549195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Dysregulation of voltage-gated sodium Na V 1.7 channels in sensory neurons contributes to chronic pain conditions, including trigeminal neuropathic pain. We previously reported that chronic pain results in part from increased SUMOylation of collapsin response mediator protein 2 (CRMP2), leading to an increased CRMP2/Na V 1.7 interaction and increased functional activity of Na V 1.7. Targeting this feed-forward regulation, we developed compound 194 , which inhibits CRMP2 SUMOylation mediated by the SUMO-conjugating enzyme Ubc9. We further demonstrated that 194 effectively reduces the functional activity of Na V 1.7 channels in dorsal root ganglia neurons and alleviated inflammatory and neuropathic pain. Here, we employed a comprehensive array of investigative approaches, encompassing biochemical, pharmacological, genetic, electrophysiological, and behavioral analyses, to assess the functional implications of Na V 1.7 regulation by CRMP2 in trigeminal ganglia (TG) neurons. We confirmed the expression of Scn9a , Dpysl2 , and UBE2I within TG neurons. Furthermore, we found an interaction between CRMP2 and Na V 1.7, with CRMP2 being SUMOylated in these sensory ganglia. Disrupting CRMP2 SUMOylation with compound 194 uncoupled the CRMP2/Na V 1.7 interaction, impeded Na V 1.7 diffusion on the plasma membrane, and subsequently diminished Na V 1.7 activity. Compound 194 also led to a reduction in TG neuron excitability. Finally, when intranasally administered to rats with chronic constriction injury of the infraorbital nerve (CCI-ION), 194 significantly decreased nociceptive behaviors. Collectively, our findings underscore the critical role of CRMP2 in regulating Na V 1.7 within TG neurons, emphasizing the importance of this indirect modulation in trigeminal neuropathic pain.
Collapse
|
5
|
Abstract
Headache disorders can produce recurrent, incapacitating pain. Migraine and cluster headache are notable for their ability to produce significant disability. The anatomy and physiology of headache disorders is fundamental to evolving treatment approaches and research priorities. Key concepts in headache mechanisms include activation and sensitization of trigeminovascular, brainstem, thalamic, and hypothalamic neurons; modulation of cortical brain regions; and activation of descending pain circuits. This review will examine the relevant anatomy of the trigeminal, brainstem, subcortical, and cortical brain regions and concepts related to the pathophysiology of migraine and cluster headache disorders.
Collapse
Affiliation(s)
- Andrea M Harriott
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yulia Orlova
- Department of Neurology, University of Florida, Gainesville, Florida
| |
Collapse
|
6
|
Dehghani A, Karatas H. Mouse Models of Familial Hemiplegic Migraine for Studying Migraine Pathophysiology. Curr Neuropharmacol 2020; 17:961-973. [PMID: 31092180 PMCID: PMC7052833 DOI: 10.2174/1570159x17666190513085013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/08/2019] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Migraine, an extremely disabling neurological disorder, has a strong genetic component. Since monogenic mi-graines (resulting from mutations or changes in a single gene) may help researchers discover migraine pathophysiology, transgenic mice models harboring gene mutations identified in Familial Hemiplegic Migraine (FHM) patients have been gen-erated. Studies in these FHM mutant mice models have shed light on the mechanisms of migraine and may aid in the identifi-cation of novel targets for treatment. More specifically, the studies shed light on how gene mutations, hormones, and other factors impact the pathophysiology of migraine. The models may also be of relevance to researchers outside the field of mi-graine as some of their aspects are relevant to pain in general. Additionally, because of the comorbidities associated with mi-graine, they share similarities with the mutant mouse models of epilepsy, stroke, and perhaps depression. Here, we review the experimental data obtained from these mutant mice and focus on how they can be used to investigate the pathophysiology of migraine, including synaptic plasticity, neuroinflammation, metabolite alterations, and molecular and behavioral mecha-nisms of pain.
Collapse
Affiliation(s)
- Anisa Dehghani
- Institute of Neurological Sciences and Psychiatry, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| |
Collapse
|
7
|
Balaban CD, Black RD, Silberstein SD. Vestibular Neuroscience for the Headache Specialist. Headache 2019; 59:1109-1127. [DOI: 10.1111/head.13550] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Carey D. Balaban
- Department of Otolaryngology University of Pittsburgh Pittsburgh PA USA
- Department of Neurobiology University of Pittsburgh Pittsburgh PA USA
- Department of Communication Sciences and Disorders University of Pittsburgh Pittsburgh PA USA
- Department of Bioengineering University of Pittsburgh Pittsburgh PA USA
| | | | - Stephen D. Silberstein
- Jefferson Headache Center, Department of Neurology Thomas Jefferson University Philadelphia PA USA
| |
Collapse
|
8
|
Abstract
Migraine is one of the world's most common neurological disorders. Current acute migraine treatments have suboptimal efficacy, and new therapeutic options are needed. Approaches targeting calcitonin gene related peptide (CGRP) signaling are clinically effective, but small molecule antagonists have not been advanced because of toxicity. In this study, we explored the axonal growth/specification collapsin response mediator protein 2 (CRMP2) as a novel “druggable” target for inhibiting CGRP release and for potential relevance for treatment of migraine pain. Collapsin response mediator protein 2 has been demonstrated to regulate N-type voltage-gated Ca2+ channel activity and Ca2+-dependent CGRP release in sensory neurons. The coexpression of CRMP2 with N-type voltage-gated Ca2+ channel and CGRP in trigeminal ganglia (TGs) sensory neurons suggested the possibility of a novel approach to regulate CGRP release in the trigeminal system. Screening protocols surprisingly revealed that (S)-lacosamide ((S)-LCM), an inactive analog of the clinically approved small molecule antiepileptic drug (R)-lacosamide (Vimpat), inhibited CRMP2 phosphorylation by cyclin-dependent kinase 5 in rat TG slices and decreased depolarization-evoked Ca2+ influx in TG cells in culture. (S)-LCM significantly blocked capsaicin-evoked CGRP release from dural nerve terminals in the rat in ex vivo cranial cup preparation. Additionally, cephalic and extracephalic cutaneous allodynia induced in rats by activation of dural nociceptors with a cocktail of inflammatory mediators, was inhibited by oral administration of (S)-LCM. The confirmation of CRMP2 as an upstream mediator of CGRP release, together with the brain penetrance of this molecule suggests (S)-LCM as a potential therapy for acute migraine.
Collapse
|
9
|
Chen SP, Tolner EA, Eikermann-Haerter K. Animal models of monogenic migraine. Cephalalgia 2016; 36:704-21. [PMID: 27154999 DOI: 10.1177/0333102416645933] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 04/01/2016] [Indexed: 01/18/2023]
Abstract
Migraine is a highly prevalent and disabling neurological disorder with a strong genetic component. Rare monogenic forms of migraine, or syndromes in which migraine frequently occurs, help scientists to unravel pathogenetic mechanisms of migraine and its comorbidities. Transgenic mouse models for rare monogenic mutations causing familial hemiplegic migraine (FHM), cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), and familial advanced sleep-phase syndrome (FASPS), have been created. Here, we review the current state of research using these mutant mice. We also discuss how currently available experimental approaches, including epigenetic studies, biomolecular analysis and optogenetic technologies, can be used for characterization of migraine genes to further unravel the functional and molecular pathways involved in migraine.
Collapse
Affiliation(s)
- Shih-Pin Chen
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taiwan Faculty of Medicine, National Yang-Ming University School of Medicine, Taiwan Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Else A Tolner
- Departments of Human Genetics and Neurology, Leiden University Medical Centre, the Netherlands
| | - Katharina Eikermann-Haerter
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| |
Collapse
|
10
|
Ferrari MD, Klever RR, Terwindt GM, Ayata C, van den Maagdenberg AMJM. Migraine pathophysiology: lessons from mouse models and human genetics. Lancet Neurol 2015; 14:65-80. [PMID: 25496898 DOI: 10.1016/s1474-4422(14)70220-0] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Migraine is a common, disabling, and undertreated episodic brain disorder that is more common in women than in men. Unbiased genome-wide association studies have identified 13 migraine-associated variants pointing at genes that cluster in pathways for glutamatergic neurotransmission, synaptic function, pain sensing, metalloproteinases, and the vasculature. The individual pathogenetic contribution of each gene variant is difficult to assess because of small effect sizes and complex interactions. Six genes with large effect sizes were identified in patients with rare monogenic migraine syndromes, in which hemiplegic migraine and non-hemiplegic migraine with or without aura are part of a wider clinical spectrum. Transgenic mouse models with human monogenic-migraine-syndrome gene mutations showed migraine-like features, increased glutamatergic neurotransmission, cerebral hyperexcitability, and enhanced susceptibility to cortical spreading depression, which is the electrophysiological correlate of aura and a putative trigger for migraine. Enhanced susceptibility to cortical spreading depression increased sensitivity to focal cerebral ischaemia, and blocking of cortical spreading depression improved stroke outcome in these mice. Changes in female hormone levels in these mice modulated cortical spreading depression susceptibility in much the same way that hormonal fluctuations affect migraine activity in patients. These findings confirm the multifactorial basis of migraine and might allow new prophylactic options to be developed, not only for migraine but potentially also for migraine-comorbid disorders such as epilepsy, depression, and stroke.
Collapse
Affiliation(s)
- Michel D Ferrari
- Department of Neurology, Leiden University Medical Centre, Leiden, Netherlands.
| | - Roselin R Klever
- Department of Human Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Centre, Leiden, Netherlands
| | - Cenk Ayata
- Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Arn M J M van den Maagdenberg
- Department of Neurology, Leiden University Medical Centre, Leiden, Netherlands; Department of Human Genetics, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
11
|
Differential trigeminovascular nociceptive responses in the thalamus in the familial hemiplegic migraine 1 knock-in mouse: A Fos protein study. Neurobiol Dis 2014; 64:1-7. [DOI: 10.1016/j.nbd.2013.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 11/15/2013] [Accepted: 12/08/2013] [Indexed: 11/16/2022] Open
|
12
|
Hoffmann J, Akerman S, Goadsby PJ. Efficacy and mechanism of anticonvulsant drugs in migraine. Expert Rev Clin Pharmacol 2014; 7:191-201. [PMID: 24494792 DOI: 10.1586/17512433.2014.885835] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Anticonvulsants represent one of the main substance classes used for the preventive treatment of migraine. Efficacy has been demonstrated in randomized placebo-controlled trials for topiramate and valproic acid including divalproex sodium. In the case of topiramate, efficacy has recently been proven for chronic migraine and even medication overuse headache, questioning the established concept of medication withdrawal. However, preventive treatment with anticonvulsants is frequently hampered by side effects that occasionally require treatment discontinuation. In addition, these data indicate that some anticonvulsant drugs are effective in migraine, while a number are clearly not useful. Effective anticonvulsants, such as topiramate and valproate, target nociceptive trigeminovascular and trigeminothalamic dural pathways or mechanisms involved in cortical spreading depression. Dissecting out how the anticonvulsants that do not work differ mechanistically from those that do will almost certainly provide avenues through which one can develop new treatments to bring to patients with migraine.
Collapse
Affiliation(s)
- Jan Hoffmann
- Headache Group-Department of Neurology, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
13
|
Akerman S, Romero-Reyes M. Insights into the pharmacological targeting of the trigeminocervical complex in the context of treatments of migraine. Expert Rev Neurother 2014; 13:1041-59. [DOI: 10.1586/14737175.2013.827472] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
14
|
Labruijere S, Ibrahimi K, Chan KY, MaassenVanDenBrink A. Discovery techniques for calcitonin gene-related peptide receptor antagonists for potential antimigraine therapies. Expert Opin Drug Discov 2013; 8:1309-23. [DOI: 10.1517/17460441.2013.826644] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
15
|
Furman JM, Marcus DA, Balaban CD. Vestibular migraine: clinical aspects and pathophysiology. Lancet Neurol 2013; 12:706-15. [DOI: 10.1016/s1474-4422(13)70107-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
16
|
Storer RJ, Goadsby PJ. Topiramate is likely to act outside of the trigeminocervical complex. Cephalalgia 2013; 33:291-300. [PMID: 23314783 DOI: 10.1177/0333102412472069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND To facilitate understanding the locus and mechanism of action of antimigraine preventives, we examined the effect of topiramate on trigeminocervical activation in the cat. METHODS Cats were anesthetized and physiologically monitored. Electrical stimulation of the superior sagittal sinus activated nociceptive trigeminovascular afferents. Extracellular recordings were made from neurons in the trigeminocervical complex. RESULTS Microiontophoretically delivered topiramate, applied locally at the second order synapse of the trigeminovascular system in the trigeminocervical complex, produced significant inhibition of L-glutamate-evoked firing of neurons only at the highest microiontophoretic currents (27 ± 7% at -160 nA; P < 0.05, N = 14 cells), but did not inhibit firing of these neurons evoked by stimulation of the craniovascular afferents (2 ± 5%, P = 0.762, N = 13 cells). In contrast, systemically administered topiramate (30 mg/kg intravenously) partly inhibited this firing (32 ± 10% at 15 min; F 5,35 = 3.5, P < 0.05, N = 8 cats). After this systemic administration, profound inhibition (70 ± 10%, P < 0.001, N = 7) of L-glutamate-evoked firing of cells in the trigeminocervical complex at the second order synapse of the trigeminovascular system was observed. CONCLUSIONS These data suggest that topiramate acts outside of the trigeminocervical complex in the cat. Determining the sites of action of preventive antimigraine treatments is crucial to developing laboratory models for the development of new therapeutics, and may vary between species.
Collapse
Affiliation(s)
- Robin J Storer
- Headache Group, Department of Neurology, UCSF Headache Center, 1701 Divisadero St., San Francisco, CA 94115, USA.
| | | |
Collapse
|
17
|
Mathew R, Andreou AP, Chami L, Bergerot A, van den Maagdenberg AMJM, Ferrari MD, Goadsby PJ. Immunohistochemical characterization of calcitonin gene-related peptide in the trigeminal system of the familial hemiplegic migraine 1 knock-in mouse. Cephalalgia 2011; 31:1368-80. [DOI: 10.1177/0333102411418847] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Introduction: Familial hemiplegic migraine type 1 (FHM-1) is caused by mutations in the CACNA1A gene, with the R192Q mutation being the most common. Elevated calcitonin gene-related peptide (CGRP) levels in acute migraine and clinical trials using CGRP receptor antagonists suggest CGRP-related mechanisms are important in migraine. Methods: Wild-type and R192Q knock-in mice were anaesthetized and perfused. Using immunohistochemical staining, the expression of CGRP in the trigeminocervical complex (TCC) and in the trigeminal and dorsal root ganglia was characterized. Results: There was a 38% reduction in the percentage of CGRP-immunoreactive cells in the trigeminal ganglia ( p < 0.001) of R192Q knock-in mice compared to wild-type animals. The size distribution profile of CGRP-immunoreactive cells within the trigeminal ganglia demonstrated no significant difference in cell diameter between the two groups ( p ≥ 0.56). CGRP expression was also reduced in thoracic ganglia of R192Q knock-in mice (21% vs. 27% in wild-type group; p < 0.05), but not in other ganglia. In addition, decreased CGRP immunoreactivity was observed in the superficial laminae of the TCC in R192Q knock-in mice, when compared to the control group ( p < 0.005). Conclusion: The data demonstrates that the FHM-1 CACNA1A mutation alters CGRP expression in the trigeminal ganglion and TCC. This suggests further study of these animals is warranted to characterize better the role of these mutations in the neurobiology of migraine.
Collapse
|
18
|
Amrutkar DV, Ploug KB, Olesen J, Jansen-Olesen I. Role for voltage gated calcium channels in calcitonin gene-related peptide release in the rat trigeminovascular system. Neuroscience 2010; 172:510-7. [PMID: 20955764 DOI: 10.1016/j.neuroscience.2010.10.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 10/08/2010] [Accepted: 10/11/2010] [Indexed: 11/16/2022]
Abstract
Clinical and genetic studies have suggested a role for voltage gated calcium channels (VGCCs) in the pathogenesis of migraine. Release of calcitonin gene-related peptide (CGRP) from trigeminal neurons has also been implicated in migraine. The VGCCs are located presynaptically on neurons and are involved in the release of these peptides to different stimuli. We have examined the presence and importance of VGCCs in controlling the CGRP release from rat dura mater, freshly isolated trigeminal ganglion (TG) and trigeminal nucleus caudalis (TNC). Each of the four VGCCs, P/Q-, N-, and L- and T-type are abundantly found in TG and TNC relative to the dura mater and each mediates a significant fraction of high potassium concentration induced CGRP release. In dura mater, blockade of P/Q-, N- and L-type VGCCs by ω-agatoxin TK, ω-conotoxin GVIA and nimodipine at 1 μM respectively, significantly decreased the potassium induced CGRP release. In the absence of calcium ions (Ca2+) and in the presence of a cocktail of blockers, the stimulated CGRP release from dura mater was reduced almost to the same level as basal CGRP release. In the TG ω-conotoxin GVIA inhibited the potassium induced CGRP release significantly. In the absence of Ca2+ and in the presence of a cocktail of blockers the stimulated CGRP release was significantly reduced. In the TNC only the cocktail of blockers and the absence of Ca2+ could reduce the potassium induced release significantly. These results suggest that depolarization by high potassium releases CGRP, and the release is regulated by Ca2+ ions and voltage-gated calcium channels.
Collapse
Affiliation(s)
- D V Amrutkar
- Department of Neurology and Danish Headache Center, Glostrup Research Institute, Glostrup Hospital, Faculty of Health Sciences, University of Copenhagen, Nordre Ringvej 69, 2600 Glostrup, Denmark
| | | | | | | |
Collapse
|
19
|
Akerman S, Holland PR, Lasalandra MP, Goadsby PJ. Inhibition of trigeminovascular dural nociceptive afferents by Ca2+-activated K+ (MaxiK/BKCa) channel opening. Pain 2010; 151:128-136. [DOI: 10.1016/j.pain.2010.06.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 06/11/2010] [Accepted: 06/26/2010] [Indexed: 11/29/2022]
|
20
|
Nair A, Simonetti M, Birsa N, Ferrari MD, van den Maagdenberg AMJM, Giniatullin R, Nistri A, Fabbretti E. Familial hemiplegic migraine Ca(v)2.1 channel mutation R192Q enhances ATP-gated P2X3 receptor activity of mouse sensory ganglion neurons mediating trigeminal pain. Mol Pain 2010; 6:48. [PMID: 20735819 PMCID: PMC2940876 DOI: 10.1186/1744-8069-6-48] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 08/24/2010] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The R192Q mutation of the CACNA1A gene, encoding for the α1 subunit of voltage-gated P/Q Ca2+ channels (Ca(v)2.1), is associated with familial hemiplegic migraine-1. We investigated whether this gain-of-function mutation changed the structure and function of trigeminal neuron P2X3 receptors that are thought to be important contributors to migraine pain. RESULTS Using in vitro trigeminal sensory neurons of a mouse genetic model knockin for the CACNA1A R192Q mutation, we performed patch clamp recording and intracellular Ca2+ imaging that showed how these knockin ganglion neurons generated P2X3 receptor-mediated responses significantly larger than wt neurons. These enhanced effects were reversed by the Ca(v)2.1 blocker ω-agatoxin. We, thus, explored intracellular signalling dependent on kinases and phosphatases to understand the molecular regulation of P2X3 receptors of knockin neurons. In such cells we observed strong activation of CaMKII reversed by ω-agatoxin treatment. The CaMKII inhibitor KN-93 blocked CaMKII phosphorylation and the hyperesponsive P2X3 phenotype. Although no significant difference in membrane expression of knockin receptors was found, serine phosphorylation of knockin P2X3 receptors was constitutively decreased and restored by KN-93. No change in threonine or tyrosine phosphorylation was detected. Finally, pharmacological inhibitors of the phosphatase calcineurin normalized the enhanced P2X3 receptor responses of knockin neurons and increased their serine phosphorylation. CONCLUSIONS The present results suggest that the CACNA1A mutation conferred a novel molecular phenotype to P2X3 receptors of trigeminal ganglion neurons via CaMKII-dependent activation of calcineurin that selectively impaired the serine phosphorylation state of such receptors, thus potentiating their effects in transducing trigeminal nociception.
Collapse
Affiliation(s)
- Asha Nair
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Andreou AP, Summ O, Charbit AR, Romero-Reyes M, Goadsby PJ. Animal models of headache: from bedside to bench and back to bedside. Expert Rev Neurother 2010; 10:389-411. [PMID: 20187862 DOI: 10.1586/ern.10.16] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In recent years bench-based studies have greatly enhanced our understanding of headache pathophysiology, while facilitating the development of new headache medicines. At present, established animal models of headache utilize activation of pain-producing cranial structures, which for a complex syndrome, such as migraine, leaves many dimensions of the syndrome unstudied. The focus on modeling the central nociceptive mechanisms and the complexity of sensory phenomena that accompany migraine may offer new approaches for the development of new therapeutics. Given the complexity of the primary headaches, multiple approaches and techniques need to be employed. As an example, recently a model for trigeminal autonomic cephalalgias has been tested successfully, while by contrast, a satisfactory model of tension-type headache has been elusive. Moreover, although useful in many regards, migraine models are yet to provide a more complete picture of the disorder.
Collapse
Affiliation(s)
- Anna P Andreou
- Headache Group - Department of Neurology, University of California, San Francisco, San Francisco, CA 94115, USA
| | | | | | | | | |
Collapse
|
22
|
GABA and valproate modulate trigeminovascular nociceptive transmission in the thalamus. Neurobiol Dis 2010; 37:314-23. [DOI: 10.1016/j.nbd.2009.10.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 10/05/2009] [Accepted: 10/09/2009] [Indexed: 11/22/2022] Open
|
23
|
Horak S, Koschak A, Stuppner H, Striessnig J. Use-dependent block of voltage-gated Cav2.1 Ca2+ channels by petasins and eudesmol isomers. J Pharmacol Exp Ther 2009; 330:220-6. [PMID: 19369579 DOI: 10.1124/jpet.109.151183] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Migraine is a frequent and often disabling disease. Treatment is unsatisfactory in many patients. A disturbed dynamic balance between excitatory and inhibitory signal processing with enhanced cortical activity probably underlies common forms of migraine. Presynaptic voltage-gated Ca(2+) channels are critical determinants of neurotransmitter release and also contribute to trigeminovascular signal transduction. Because clinical evidence exists for migraine-prophylactic actions of Petasites hybridus extracts, we investigated whether petasins comprising the main constituents of the extract inhibit currents through presynaptic Ca(v)2.1 channels expressed in Xenopus laevis oocytes. P. hybridus extract (0.02 mg/ml), petasin, neopetasin, isopetasin, S-petasin, and iso-S-petasin (50 microM) were weak tonic blockers of Ca(v)2.1-mediated barium currents (I(Ba)) during infrequent depolarizations (0.1 Hz), but their inhibitory potency increased at higher stimulation rates (1 Hz), indicating preferential block of open and/or inactivated channels. Sulfur-containing compounds (S-petasin, Iso-S-petasin) were the most potent significantly promoting the accumulation of Ca(v)2.1 channel in inactivated states during pulse trains (I(Ba) decrease during 1-Hz pulse trains: control, 45%, S-petasin, 79%; iso-S-petasin, 80%). For the Eucalyptus williamsiania sesquiterpenes alpha- and gamma-eudesmol, a comparable use-dependent inhibition was found in addition to a tonic block component. Alpha-eudesmol and petasins accelerated the voltage-dependent inactivation of Ca(v)2.1 channels during depolarizations. We demonstrate that S-petasin, iso-S-petasin, and eudesmol are Ca(v)2.1 channel inhibitors preferentially acting as use-dependent channel blockers and with the sulfur-containing substituent in position 3 of the petasins serving as important functional feature. The Ca(v)2.1-inhibitory properties of these petasins may contribute to migraine-prophylactic properties described for P. hybridus extracts.
Collapse
Affiliation(s)
- Silja Horak
- Institute of Pharmacy, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria
| | | | | | | |
Collapse
|
24
|
Martella G, Costa C, Pisani A, Cupini LM, Bernardi G, Calabresi P. Antiepileptic Drugs on Calcium Currents Recorded from Cortical and PAG Neurons: Therapeutic Implications for Migraine. Cephalalgia 2008; 28:1315-26. [DOI: 10.1111/j.1468-2982.2008.01682.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cortex and periaqueductal grey (PAG) play a major role in the pathophysiology of migraine. Some antiepileptic drugs (AEDs) influence the activity of these structures by modulating high-voltage-activated (HVA) Ca2+ channels and are effective in migraine prevention. The aim of the present study was to investigate the expression of total HVA Ca2+ channels in cortical and PAG neurons and to study the differential action of AEDs on these channels. Isolated neurons were visually identified based on morphological criteria. HVA currents were recorded by whole-cell patch-clamp technique. The distribution ratio of L-, N-, P-, Q- and R-type HVA Ca2+ channels was different between cortical and PAG neurons. In particular, we found that P- and Q-type HVA Ca2+ channels were more expressed in PAG neurons than in cortical cells, whereas L- and R-type HVA Ca2+ channels showed an opposite distribution. Interestingly, N-type HVA Ca2+ channels were equally distributed in these two neuronal populations. A differential sensitivity to AEDs of HVA Ca2+ channels located on cortical and PAG neurons was observed for topiramate (TPM), but not for lamotrigine (LTG) or levetiracetam (LEV). In fact, whereas both LTG and LEV were equally effective and potent in inhibiting HVA Ca2+ currents in the two neuronal populations, TPM showed a much higher potency and efficacy in blocking these currents in PAG neurons than in cortical pyramidal cells. TPM, in fact, inhibited N-, P- and L-type channels in PAG neurons, whereas in cortical neurons this AED modulated only P- and L-type channels. Unlike the other AEDs investigated, valproic acid did not affect HVA Ca2+ currents in cortical and PAG neurons. The negative modulation of specific subtypes of HVA Ca2+ channels by various AEDs can restore normal electrical activity in target brain areas such as cortex and PAG, providing interesting therapeutic approaches in migraine prevention.
Collapse
Affiliation(s)
- G Martella
- Clinica Neurologica, Dipartimento di Neuroscienze, Università di Roma ‘Tor Vergata’
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS c/o CERC, Rome
| | - C Costa
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS c/o CERC, Rome
- Clinica Neurologica, Dip. Specialità Medico Chirurgiche e Sanità Pubblica, Università di Perugia, Ospedale Santa Maria della Misericordia, Perugia
| | - A Pisani
- Clinica Neurologica, Dipartimento di Neuroscienze, Università di Roma ‘Tor Vergata’
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS c/o CERC, Rome
| | - LM Cupini
- Cinica Neurologica, Ospedale S. Eugenio, Rome, Italy
| | - G Bernardi
- Clinica Neurologica, Dipartimento di Neuroscienze, Università di Roma ‘Tor Vergata’
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS c/o CERC, Rome
| | - P Calabresi
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS c/o CERC, Rome
- Clinica Neurologica, Dip. Specialità Medico Chirurgiche e Sanità Pubblica, Università di Perugia, Ospedale Santa Maria della Misericordia, Perugia
| |
Collapse
|
25
|
Vanmolkot KRJ, Terwindt GM, Frants RR, Haan J, van den Maagdenberg AMJM, Ferrari MD. A Gene for a New Monogenic Neurovascular Migraine Syndrome: A Next Step in Unravelling Molecular Pathways for Migraine? Cephalalgia 2008; 28:471-3. [DOI: 10.1111/j.1468-2982.2007.01511.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- KRJ Vanmolkot
- Department of Human Genetics, Leiden University Medical Centre, Leiden
| | - GM Terwindt
- Department of Neurology, Leiden University Medical Centre, Leiden
| | - RR Frants
- Department of Human Genetics, Leiden University Medical Centre, Leiden
| | - J Haan
- Department of Neurology, Leiden University Medical Centre, Leiden
- Department of Neurology, Rijnland Hospital, Leiderdorp, the Netherlands
| | - AMJM van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Centre, Leiden
- Department of Neurology, Leiden University Medical Centre, Leiden
| | - MD Ferrari
- Department of Neurology, Leiden University Medical Centre, Leiden
| |
Collapse
|
26
|
Xiao Y, Richter JA, Hurley JH. Release of glutamate and CGRP from trigeminal ganglion neurons: Role of calcium channels and 5-HT1 receptor signaling. Mol Pain 2008; 4:12. [PMID: 18416824 PMCID: PMC2359740 DOI: 10.1186/1744-8069-4-12] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 04/16/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aberrant release of the neurotransmitters, glutamate and calcitonin-gene related peptide (CGRP), from trigeminal neurons has been implicated in migraine. The voltage-gated P/Q-type calcium channel has a critical role in controlling neurotransmitter release and has been linked to Familial Hemiplegic Migraine. Therefore, we examined the importance of voltage-dependent calcium channels in controlling release of glutamate and CGRP from trigeminal ganglion neurons isolated from male and female rats and grown in culture. Serotonergic pathways are likely involved in migraine, as triptans, a class of 5-HT1 receptor agonists, are effective in the treatment of migraine and their effectiveness may be due to inhibiting neurotransmitter release from trigeminal neurons. We also studied the effect of serotonin receptor activation on release of glutamate and CGRP from trigeminal neurons grown in culture. RESULTS P/Q-, N- and L-type channels each mediate a significant fraction of potassium-stimulated release of glutamate and CGRP. We determined that 5-HT significantly inhibits potassium-stimulated release of both glutamate and CGRP. Serotonergic inhibition of both CGRP and glutamate release can be blocked by pertussis toxin and NAS-181, a 5-HT1B/1D antagonist. Stimulated release of CGRP is unaffected by Y-25130, a 5-HT3 antagonist and SB 200646, a 5-HT2B/2C antagonist. CONCLUSION These data suggest that release of both glutamate and CGRP from trigeminal neurons is controlled by calcium channels and modulated by 5-HT signaling in a pertussis-toxin dependent manner and probably via 5-HT1 receptor signaling. This is the first characterization of glutamate release from trigeminal neurons grown in culture.
Collapse
Affiliation(s)
- Yan Xiao
- Department of Biochemistry and Molecular Biology, Stark Neurosciences Research Institute, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, Indiana, 46202, USA.
| | | | | |
Collapse
|
27
|
Shank RP, Maryanoff BE. Molecular pharmacodynamics, clinical therapeutics, and pharmacokinetics of topiramate. CNS Neurosci Ther 2008; 14:120-42. [PMID: 18482025 PMCID: PMC6494007 DOI: 10.1111/j.1527-3458.2008.00041.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Topiramate (TPM; TOPAMAX) is a broad-spectrum antiepileptic drug (AED) that is approved in many world markets for preventing or reducing the frequency of epileptic seizures (as monotherapy or adjunctive therapy), and for the prophylaxis of migraine. TPM, a sulfamate derivative of the naturally occurring sugar D-fructose, possesses several pharmacodynamic properties that may contribute to its clinically useful attributes, and to its observed adverse effects. The sulfamate moiety is essential, but not sufficient, for its pharmacodynamic properties. In this review, we discuss the known pharmacodynamic and pharmacokinetic properties of TPM, as well as its various clinically beneficial and adverse effects.
Collapse
Affiliation(s)
- Richard P. Shank
- Research & Early Development, Johnson & Johnson Pharmaceutical Research & Development, Spring House, Pennsylvania 19477‐0776 USA
| | - Bruce E. Maryanoff
- Research & Early Development, Johnson & Johnson Pharmaceutical Research & Development, Spring House, Pennsylvania 19477‐0776 USA
| |
Collapse
|
28
|
Todorov B, van de Ven RCG, Kaja S, Broos LAM, Verbeek SJ, Plomp JJ, Ferrari MD, Frants RR, van den Maagdenberg AMJM. Conditional inactivation of the Cacna1a gene in transgenic mice. Genesis 2007; 44:589-94. [PMID: 17146767 DOI: 10.1002/dvg.20255] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ca(v)2.1 (P/Q-type) voltage-gated calcium channels play an important role in neurotransmitter release at many brain synapses and at the neuromuscular junction. Mutations in the CACNA1A gene, encoding the pore forming alpha(1) subunit of Ca(v)2.1 channels, are associated with a wide spectrum of neurological disorders. Here we generated mice with a conditional, floxed, Cacna1a allele without any overt phenotype. Deletion of the floxed Cacna1a allele resulted in ataxia, dystonia, and lethality during the fourth week, a severe phenotype similar to conventional Ca(v)2.1 knockout mice. Although neurotransmitter release at the neuromuscular junction was not affected in the conditional mice, homozygous deletion of the floxed allele caused an ablation of Ca(v)2.1 channel-mediated neurotransmission that was accompanied by a compensatory upregulation of Ca(v)2.3 (R-type) channels at this synapse. Pharmacological inhibition of Ca(v)2.1 channels is possible, but the contributing cell-types and time windows relevant to the different Ca(v)2.1-related neurological disorders can only be reliably determined using Cacna1a conditional mice.
Collapse
Affiliation(s)
- Boyan Todorov
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Migraine as a Cerebral Ionopathy with Abnormal Central Sensory Processing**Some portions of the pathophysiology have appeared fully referenced [1]. Neurobiol Dis 2007. [DOI: 10.1016/b978-012088592-3/50034-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] Open
|
30
|
Abstract
Serotonin (5-HT)(1B/1D) receptor agonists, which are also known as triptans, represent the most important advance in migraine therapeutics in the four millennia that the condition has been recognized. The vasoconstrictive activity of triptans produced a small clinical penalty in terms of coronary vasoconstriction but also raised an enormous intellectual question: to what extent is migraine a vascular problem? Functional neuroimaging and neurophysiological studies have consistently developed the theme of migraine as a brain disorder and, therefore, demanded that the search for neurally acting antimigraine drugs should be undertaken. The prospect of non-vasoconstrictor acute migraine therapies, potential targets for which are discussed here, offers a real opportunity to patients and provides a therapeutic rationale that places migraine firmly in the brain as a neurological problem, where it undoubtedly belongs.
Collapse
Affiliation(s)
- Peter J Goadsby
- Headache Group, Institute of Neurology, and The National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
31
|
Hoebeek FE, Stahl JS, van Alphen AM, Schonewille M, Luo C, Rutteman M, van den Maagdenberg AMJM, Molenaar PC, Goossens HHLM, Frens MA, De Zeeuw CI. Increased noise level of purkinje cell activities minimizes impact of their modulation during sensorimotor control. Neuron 2005; 45:953-65. [PMID: 15797555 DOI: 10.1016/j.neuron.2005.02.012] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2004] [Revised: 09/27/2004] [Accepted: 02/02/2005] [Indexed: 11/24/2022]
Abstract
While firing rate is well established as a relevant parameter for encoding information exchanged between neurons, the significance of other parameters is more conjectural. Here, we show that regularity of neuronal spike activities affects sensorimotor processing in tottering mutants, which suffer from a mutation in P/Q-type voltage-gated calcium channels. While the modulation amplitude of the simple spike firing rate of their floccular Purkinje cells during optokinetic stimulation is indistinguishable from that of wild-types, the regularity of their firing is markedly disrupted. The gain and phase values of tottering's compensatory eye movements are indistinguishable from those of flocculectomized wild-types or from totterings with the flocculus treated with P/Q-type calcium channel blockers. Moreover, normal eye movements can be evoked in tottering when the flocculus is electrically stimulated with regular spike trains mimicking the firing pattern of normal simple spikes. This study demonstrates the importance of regularity of firing in Purkinje cells for neuronal information processing.
Collapse
Affiliation(s)
- F E Hoebeek
- Department of Neuroscience, Erasmus MC, Dr. Molenwaterplein 50, P.O. Box 1738, 3000 DR Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|