1
|
Cavalcante DP, Nunes AÍDS, da Silva ER, de Carvalho GA, Chiareli RA, Oliveira-Lima OC, Ortiz-Leoncini G, Ulrich H, Gomez RS, Pinto MCX. GlyT1 inhibition promotes neuroprotection in the middle cerebral artery occlusion model through the activation of GluN2A-containing NMDAR. Exp Neurol 2024; 383:115006. [PMID: 39424040 DOI: 10.1016/j.expneurol.2024.115006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Glycine Transporter Type 1 (GlyT1) inhibition confers neuroprotection against different forms of cerebral damage. This effect occurs through the elevation of synaptic glycine concentrations, which enhances N-methyl-d-aspartate receptor (NMDAR) activation by glutamate. To investigate the neuroprotective mechanism of GlyT1 inhibition, we used the Middle Cerebral Artery Occlusion (MCAO) model in male C57BL/6 mice, aged 10-12 weeks. We administered N-[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl] sarcosine (NFPS), a GlyT1 inhibitor, 24 h prior to ischemia induction. NFPS pretreatment provided significant neuroprotection in the MCAO model, associated with modulation of pathways related to long-term potentiation. Specifically, GluN2A subunit expression was upregulated, while GluN2B subunit expression was downregulated in cortical areas, correlating with enhanced phosphorylation of CaMKIV and CREB proteins. Coadministration with the GluN2B antagonist Eliprodil or the CREB inhibitor C646 did not affect the neuroprotective effects of NFPS pretreatment, but TCN-201, a specific GluN2A antagonist, disrupted these effects. These findings suggest that GlyT1 inhibition mediates neuroprotection through activation of GluN2A-containing NMDARs and the GluN2A/CaMKIV/CREB signaling cascade, thereby modulating the balance between GluN2A and GluN2B subunits.
Collapse
Affiliation(s)
- Daniel Pereira Cavalcante
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goias, Goiânia, GO, Brazil
| | | | - Eduardo Rosa da Silva
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goias, Goiânia, GO, Brazil
| | - Gustavo Almeida de Carvalho
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goias, Goiânia, GO, Brazil
| | - Raphaela Almeida Chiareli
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goias, Goiânia, GO, Brazil
| | | | - Giovanni Ortiz-Leoncini
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goias, Goiânia, GO, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Renato Santiago Gomez
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro Cunha Xavier Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goias, Goiânia, GO, Brazil.
| |
Collapse
|
2
|
Li H, Wang C, Gong Z, Nie L, Xu J, Wang M. Transient Receptor Potential Ankyrin 1-dependent Activation of Extracellular Signal-regulated Kinase 2 in the Cerebral Cortices Contributes to Cortical Spreading Depolarization. Neuroscience 2024; 543:90-100. [PMID: 38417540 DOI: 10.1016/j.neuroscience.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 03/01/2024]
Abstract
Extracellular signal-regulated kinase (ERK) are serine/threonine-selective proteins and ERK1/2 can be phosphorylated in peripheral and central brain regions after cortical spreading depolarization (CSD) and calcitonin gene-related peptide; However, it remains unclear about whether and how ERK activity modulates CSD that correlates to migraine aura. Here, we determined the role of ERK in regulating CSD and explored the underlying mechanism involving transient receptor potential ankyrin 1 (TRPA1), a stress-sensing cation channel. CSD was recorded using intrinsic optical imaging in mouse brain slices, and electrophysiology in rats. Phosphorylated ERK (pERK1/2) and interleukin-1β (IL-1β) protein levels were detected using Western blot or enzyme-linked immunosorbent assay, respectively. IL-1β mRNA level was detected using qPCR. The results showed that an ERK inhibitor, SCH77298, markedly prolonged CSD latency and reduced propagation rate in mouse brain slices. Corresponding to this, CSD induction increased levels of cytosolic pERK1/2 in ipsilateral cerebral cortices of rats, the elevation of which correlated to the level of IL-1β mRNA. Mechanistic analysis showed that pre-treatment of an anti-TRPA1 antibody reduced the cytosolic pERK2 level but not pERK1 following CSD in cerebral cortices of rats and this level of pERK2 correlated with that of cerebral cortical IL-1β protein. Furthermore, an ERK activator, AES16-2M, but not its scrambled control, reversed the prolonged CSD latency by a TRPA1 inhibitor, HC-030031, in mouse brain slices. These data revealed a crucial role of ERK activity in regulating CSD, and elevation of pERK and IL-1β production induced by CSD is predominantly TRPA1 channel-dependent, thereby contributing to migraine pathogenesis.
Collapse
Affiliation(s)
- Haoyang Li
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Chenyi Wang
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Ziyang Gong
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Lingdi Nie
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Jiaxin Xu
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Minyan Wang
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China.
| |
Collapse
|
3
|
Liu R, Zhang S, Zeng H, Gao N, Yin Y, Zhang M, Mao L. A Potentiometric Dual-Channel Microsensor Reveals that Fluctuation of H 2 S is Less pH-Dependent During Spreading Depolarization in the Rat Brain. Angew Chem Int Ed Engl 2024; 63:e202318973. [PMID: 38272831 DOI: 10.1002/anie.202318973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 01/27/2024]
Abstract
Spreading depolarization (SD) is one of the most common neuropathologic phenomena in the nervous system, relating to numerous diseases. However, real-time monitoring the rapid chemical changes during SD to probe the molecular mechanism remains a great challenge. We develop a potentiometric dual-channel microsensor for simultaneous monitoring of H2 S and pH featuring excellent selectivity and spatiotemporal resolution. Using this microsensor we first observe real time changes of H2 S and pH in the rat brain induced by SD. This changes of H2 S are completely suppressed when the rat pre-treats with aminooxyacetic acid (AOAA), a blocker to inhibit the H2 S-producing enzyme, indicating H2 S fluctuation might be related to enzyme-dependent pathway during SD and less pH-dependent. This study provides a new perspective for studying the function of H2 S and the molecular basis of SD-associated diseases.
Collapse
Affiliation(s)
- Rantong Liu
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing, 100872, China
- College of Petroleum and Environment Engineering, Yan'an University, Shaanxi Yan'an, 716000, China
| | - Shuai Zhang
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing, 100872, China
| | - Hui Zeng
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing, 100872, China
| | - Nan Gao
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing, 100872, China
| | - Yongyue Yin
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing, 100872, China
| | - Meining Zhang
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing, 100872, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
4
|
Viudez-Martínez A, Torregrosa AB, Navarrete F, García-Gutiérrez MS. Understanding the Biological Relationship between Migraine and Depression. Biomolecules 2024; 14:163. [PMID: 38397400 PMCID: PMC10886628 DOI: 10.3390/biom14020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.
Collapse
Affiliation(s)
- Adrián Viudez-Martínez
- Hospital Pharmacy Service, Hospital General Dr. Balmis de Alicante, 03010 Alicante, Spain;
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
5
|
Sgro M, Ray J, Foster E, Mychasiuk R. Making migraine easier to stomach: the role of the gut-brain-immune axis in headache disorders. Eur J Neurol 2023; 30:3605-3621. [PMID: 37329292 DOI: 10.1111/ene.15934] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND AND PURPOSE Headache disorders place a significant burden on the healthcare system, being the leading cause of disability in those under 50 years. Novel studies have interrogated the relationship between headache disorders and gastrointestinal dysfunction, suggesting a link between the gut-brain-immune (GBI) axis and headache pathogenesis. Although the exact mechanisms driving the complex relationship between the GBI axis and headache disorders remain unclear, there is a growing appreciation that a healthy and diverse microbiome is necessary for optimal brain health. METHODS A literature search was performed through multiple reputable databases in search of Q1 journals within the field of headache disorders and gut microbiome research and were critically and appropriately evaluated to investigate and explore the following; the role of the GBI axis in dietary triggers of headache disorders and the evidence indicating that diet can be used to alleviate headache severity and frequency. The relationship between the GBI axis and post-traumatic headache is then synthesized. Finally, the scarcity of literature regarding paediatric headache disorders and the role that the GBI axis plays in mediating the relationship between sex hormones and headache disorders are highlighted. CONCLUSIONS There is potential for novel therapeutic targets for headache disorders if understanding of the GBI axis in their aetiology, pathogenesis and recovery is increased.
Collapse
Affiliation(s)
- Marissa Sgro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jason Ray
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Austin Health, Melbourne, Victoria, Australia
| | - Emma Foster
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Cao J, Grover P, Kainerstorfer JM. A model of neurovascular coupling and its application to cortical spreading depolarization. J Theor Biol 2023; 572:111580. [PMID: 37459953 DOI: 10.1016/j.jtbi.2023.111580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/09/2023] [Accepted: 07/10/2023] [Indexed: 08/01/2023]
Abstract
Cortical spreading depolarization (CSD) is a neuropathological condition involving propagating waves of neuronal silence, and is related to multiple diseases, such as migraine aura, traumatic brain injury (TBI), stroke, and cardiac arrest, as well as poor outcome of patients. While CSDs of different severity share similar roots on the ion exchange level, they can lead to different vascular responses (namely spreading hyperemia and spreading ischemia). In this paper, we propose a mathematical model relating neuronal activities to predict vascular changes as measured with near-infrared spectroscopy (NIRS) and fMRI recordings, and apply it to the extreme case of CSD, where sustained near-complete neuronal depolarization is seen. We utilize three serially connected models (namely, ion exchange, neurovascular coupling, and hemodynamic model) which are described by differential equations. Propagating waves of ion concentrations, as well as the associated vasodynamics and hemodynamics, are simulated by solving these equations. Our proposed model predicts vasodynamics and hemodynamics that agree both qualitatively and quantitatively with experimental literature. Mathematical modeling and simulation offer a powerful tool to help understand the underlying mechanisms of CSD and help interpret the data. In addition, it helps develop novel monitoring techniques prior to data collection. Our simulated results strongly suggest that fMRI is unable to reliably distinguish between spreading hyperemia and spreading ischemia, while NIRS signals are substantially distinct in the two cases.
Collapse
Affiliation(s)
- Jiaming Cao
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, 15213, PA, United States
| | - Pulkit Grover
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, 15213, PA, United States; Department of Electrical and Computer Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, 15213, PA, United States; Neuroscience Institute, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, 15213, PA, United States
| | - Jana M Kainerstorfer
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, 15213, PA, United States; Department of Electrical and Computer Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, 15213, PA, United States; Neuroscience Institute, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, 15213, PA, United States.
| |
Collapse
|
7
|
Amani H, Soltani Khaboushan A, Terwindt GM, Tafakhori A. Glia Signaling and Brain Microenvironment in Migraine. Mol Neurobiol 2023; 60:3911-3934. [PMID: 36995514 DOI: 10.1007/s12035-023-03300-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/27/2023] [Indexed: 03/31/2023]
Abstract
Migraine is a complicated neurological disorder affecting 6% of men and 18% of women worldwide. Various mechanisms, including neuroinflammation, oxidative stress, altered mitochondrial function, neurotransmitter disturbances, cortical hyperexcitability, genetic factors, and endocrine system problems, are responsible for migraine. However, these mechanisms have not completely delineated the pathophysiology behind migraine, and they should be further studied. The brain microenvironment comprises neurons, glial cells, and vascular structures with complex interactions. Disruption of the brain microenvironment is the main culprit behind various neurological disorders. Neuron-glia crosstalk contributes to hyperalgesia in migraine. In the brain, microenvironment and related peripheral regulatory circuits, microglia, astrocytes, and satellite cells are necessary for proper function. These are the most important cells that could induce migraine headaches by disturbing the balance of the neurotransmitters in the nervous system. Neuroinflammation and oxidative stress are the prominent reactions glial cells drive during migraine. Understanding the role of cellular and molecular components of the brain microenvironment on the major neurotransmitters engaged in migraine pathophysiology facilitates the development of new therapeutic approaches with higher effectiveness for migraine headaches. Investigating the role of the brain microenvironment and neuroinflammation in migraine may help decipher its pathophysiology and provide an opportunity to develop novel therapeutic approaches for its management. This review aims to discuss the neuron-glia interactions in the brain microenvironment during migraine and their potential role as a therapeutic target for the treatment of migraine.
Collapse
Affiliation(s)
- Hanieh Amani
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Soltani Khaboushan
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Neurology, Imam Khomeini Hospital, Keshavarz Blvd., Tehran, Iran.
| |
Collapse
|
8
|
Kalatharan V, Al-Karagholi MAM. Targeting Peripheral N-Methyl-D-Aspartate Receptor (NMDAR): A Novel Strategy for the Treatment of Migraine. J Clin Med 2023; 12:jcm12062156. [PMID: 36983158 PMCID: PMC10055974 DOI: 10.3390/jcm12062156] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Backgrounds: Several acute and preventive medications were developed for the treatment of migraine. Yet, a significant proportion of patients reports an inadequate response and a lack of tolerability, emphasizing the need for new options. Glutamate is the most important excitatory neurotransmitter in the brain, and glutamate receptors including N-Methyl-D-Aspartate Receptor (NMDAR) are expressed at several levels of the trigeminovascular system, which is the anatomical and physiological substrate of migraine pain. Objective: To review preclinical and clinical studies investigating the role of the NMDAR in migraine pathophysiology. Methods: No protocol was registered for this study. References for the present review were identified from a narrative search of the PubMed database. Search terms such as glutamate, migraine, N-Methyl-D-Aspartate Receptor, and NMDAR were used. No restrictions were made in terms of the language and date of publication. Results: In animal models, administration of monosodium glutamate (MSG) activated and sensitized trigeminovascular neurons. In healthy human participants, consumption of MSG caused headaches, craniofacial sensitivity, and nausea. In in vivo models and through immunolabeling, NMDAR subunits NR1, NR2A, and NR2B were expressed in trigeminal ganglion neurons. In humans, NMDAR antagonists such as ketamine and memantine caused a significant reduction in pain intensity and monthly headache frequency. Conclusions: Accumulative evidence indicates that NMDAR is a promising new target for the treatment of migraine. Selective NMDAR antagonists without central effects are needed to investigate their therapeutic benefit in the treatment of migraine.
Collapse
|
9
|
Giniatullin R, Nistri A. Role of ATP in migraine mechanisms: focus on P2X3 receptors. J Headache Pain 2023; 24:1. [PMID: 36597043 PMCID: PMC9809127 DOI: 10.1186/s10194-022-01535-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/16/2022] [Indexed: 01/04/2023] Open
Abstract
Migraine is a major health burden worldwide with complex pathophysiology and multifarious underlying mechanisms. One poorly understood issue concerns the early steps in the generation of migraine pain. To elucidate the basic process of migraine pain further, it seems useful to consider key molecular players that may operate synergistically to evoke headache. While the neuropeptide CGRP is an important contributor, we propose that extracellular ATP (that generally plays a powerful nociceptive role) is also a major component of migraine headache, acting in concert with CGRP to stimulate trigeminal nociceptive neurons. The aim of the present focused review is to highlight the role of ATP activating its P2X3 membrane receptors selectively expressed by sensory neurons including their nerve fiber terminals in the meninges. Specifically, we present data on the homeostasis of ATP and related purines in the trigeminovascular system and in the CNS; the basic properties of ATP signalling at peripheral and central nerve terminals; the characteristics of P2X3 and related receptors in trigeminal neurons; the critical speed and persistence of P2X3 receptor activity; their cohabitation at the so-called meningeal neuro-immune synapse; the identity of certain endogenous agents cooperating with ATP to induce neuronal sensitization in the trigeminal sensory system; the role of P2X3 receptors in familial type migraine; the current state of P2X3 receptor antagonists and their pharmacological perspectives in migraine. It is proposed that the unique kinetic properties of P2X3 receptors activated by ATP offer an interesting translational value to stimulate future studies for innovative treatments of migraine pain.
Collapse
Affiliation(s)
- R. Giniatullin
- grid.9668.10000 0001 0726 2490A.I Virtanen Institute, University of Eastern Finland, 70211 Kuopio, Finland
| | - A. Nistri
- grid.5970.b0000 0004 1762 9868Department of Neuroscience, International School for Advanced Studies (SISSA), 34136 Trieste, Italy
| |
Collapse
|
10
|
Inhibiting Endocannabinoid Hydrolysis as Emerging Analgesic Strategy Targeting a Spectrum of Ion Channels Implicated in Migraine Pain. Int J Mol Sci 2022; 23:ijms23084407. [PMID: 35457225 PMCID: PMC9027089 DOI: 10.3390/ijms23084407] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 12/23/2022] Open
Abstract
Migraine is a disabling neurovascular disorder characterized by severe pain with still limited efficient treatments. Endocannabinoids, the endogenous painkillers, emerged, alternative to plant cannabis, as promising analgesics against migraine pain. In this thematic review, we discuss how inhibition of the main endocannabinoid-degrading enzymes, monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), could raise the level of endocannabinoids (endoCBs) such as 2-AG and anandamide in order to alleviate migraine pain. We describe here: (i) migraine pain signaling pathways, which could serve as specific targets for antinociception; (ii) a divergent distribution of MAGL and FAAH activities in the key regions of the PNS and CNS implicated in migraine pain signaling; (iii) a complexity of anti-nociceptive effects of endoCBs mediated by cannabinoid receptors and through a direct modulation of ion channels in nociceptive neurons; and (iv) the spectrum of emerging potent MAGL and FAAH inhibitors which efficiently increase endoCBs levels. The specific distribution and homeostasis of endoCBs in the main regions of the nociceptive system and their generation ‘on demand’, along with recent availability of MAGL and FAAH inhibitors suggest new perspectives for endoCBs-mediated analgesia in migraine pain.
Collapse
|
11
|
Guerrero-Toro C, Koroleva K, Ermakova E, Gafurov O, Abushik P, Tavi P, Sitdikova G, Giniatullin R. Testing the Role of Glutamate NMDA Receptors in Peripheral Trigeminal Nociception Implicated in Migraine Pain. Int J Mol Sci 2022; 23:ijms23031529. [PMID: 35163452 PMCID: PMC8835926 DOI: 10.3390/ijms23031529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 01/18/2023] Open
Abstract
The pro-nociceptive role of glutamate in the CNS in migraine pathophysiology is well established. Glutamate, released from trigeminal afferents, activates second order nociceptive neurons in the brainstem. However, the function of peripheral glutamate receptors in the trigeminovascular system suggested as the origin site for migraine pain, is less known. In the current project, we used calcium imaging and patch clamp recordings from trigeminal ganglion (TG) neurons, immunolabelling, CGRP assay and direct electrophysiological recordings from rat meningeal afferents to investigate the role of glutamate in trigeminal nociception. Glutamate, aspartate, and, to a lesser extent, NMDA under free-magnesium conditions, evoked calcium transients in a fraction of isolated TG neurons, indicating functional expression of NMDA receptors. The fraction of NMDA sensitive neurons was increased by the migraine mediator CGRP. NMDA also activated slowly desensitizing currents in 37% of TG neurons. However, neither glutamate nor NMDA changed the level of extracellular CGRP. TG neurons expressed both GluN2A and GluN2B subunits of NMDA receptors. In addition, after removal of magnesium, NMDA activated persistent spiking activity in a fraction of trigeminal nerve fibers in meninges. Thus, glutamate activates NMDA receptors in somas of TG neurons and their meningeal nerve terminals in magnesium-dependent manner. These findings suggest that peripherally released glutamate can promote excitation of meningeal afferents implicated in generation of migraine pain in conditions of inherited or acquired reduced magnesium blockage of NMDA channels and support the usage of magnesium supplements in migraine.
Collapse
Affiliation(s)
- Cindy Guerrero-Toro
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (C.G.-T.); (K.K.); (P.A.); (P.T.)
| | - Kseniia Koroleva
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (C.G.-T.); (K.K.); (P.A.); (P.T.)
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.E.); (O.G.)
| | - Elizaveta Ermakova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.E.); (O.G.)
| | - Oleg Gafurov
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.E.); (O.G.)
| | - Polina Abushik
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (C.G.-T.); (K.K.); (P.A.); (P.T.)
- Laboratory of Comparative Neurophysiology, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 Saint Petersburg, Russia
| | - Pasi Tavi
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (C.G.-T.); (K.K.); (P.A.); (P.T.)
| | - Guzel Sitdikova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.E.); (O.G.)
- Correspondence: (G.S.); (R.G.); Tel.: +7-9033061092 (G.S.); +358-403553665 (R.G.)
| | - Rashid Giniatullin
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (C.G.-T.); (K.K.); (P.A.); (P.T.)
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.E.); (O.G.)
- Correspondence: (G.S.); (R.G.); Tel.: +7-9033061092 (G.S.); +358-403553665 (R.G.)
| |
Collapse
|
12
|
Mathew AA, Panonnummal R. Cortical spreading depression: culprits and mechanisms. Exp Brain Res 2022; 240:733-749. [DOI: 10.1007/s00221-022-06307-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 01/06/2022] [Indexed: 02/14/2023]
|
13
|
Demchenko I, Tassone VK, Kennedy SH, Dunlop K, Bhat V. Intrinsic Connectivity Networks of Glutamate-Mediated Antidepressant Response: A Neuroimaging Review. Front Psychiatry 2022; 13:864902. [PMID: 35722550 PMCID: PMC9199367 DOI: 10.3389/fpsyt.2022.864902] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/12/2022] [Indexed: 11/23/2022] Open
Abstract
Conventional monoamine-based pharmacotherapy, considered the first-line treatment for major depressive disorder (MDD), has several challenges, including high rates of non-response. To address these challenges, preclinical and clinical studies have sought to characterize antidepressant response through monoamine-independent mechanisms. One striking example is glutamate, the brain's foremost excitatory neurotransmitter: since the 1990s, studies have consistently reported altered levels of glutamate in MDD, as well as antidepressant effects following molecular targeting of glutamatergic receptors. Therapeutically, this has led to advances in the discovery, testing, and clinical application of a wide array of glutamatergic agents, particularly ketamine. Notably, ketamine has been demonstrated to rapidly improve mood symptoms, unlike monoamine-based interventions, and the neurobiological basis behind this rapid antidepressant response is under active investigation. Advances in brain imaging techniques, including functional magnetic resonance imaging, magnetic resonance spectroscopy, and positron emission tomography, enable the identification of the brain network-based characteristics distinguishing rapid glutamatergic modulation from the effect of slow-acting conventional monoamine-based pharmacology. Here, we review brain imaging studies that examine brain connectivity features associated with rapid antidepressant response in MDD patients treated with glutamatergic pharmacotherapies in contrast with patients treated with slow-acting monoamine-based treatments. Trends in recent brain imaging literature suggest that the activity of brain regions is organized into coherent functionally distinct networks, termed intrinsic connectivity networks (ICNs). We provide an overview of major ICNs implicated in depression and explore how treatment response following glutamatergic modulation alters functional connectivity of limbic, cognitive, and executive nodes within ICNs, with well-characterized anti-anhedonic effects and the enhancement of "top-down" executive control. Alterations within and between the core ICNs could potentially exert downstream effects on the nodes within other brain networks of relevance to MDD that are structurally and functionally interconnected through glutamatergic synapses. Understanding similarities and differences in brain ICNs features underlying treatment response will positively impact the trajectory and outcomes for adults suffering from MDD and will facilitate the development of biomarkers to enable glutamate-based precision therapeutics.
Collapse
Affiliation(s)
- Ilya Demchenko
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Center for Depression and Suicide Studies, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Vanessa K Tassone
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Sidney H Kennedy
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Center for Depression and Suicide Studies, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Katharine Dunlop
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Center for Depression and Suicide Studies, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Venkat Bhat
- Interventional Psychiatry Program, Mental Health and Addictions Service, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Center for Depression and Suicide Studies, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Vuralli D, Karatas H, Yemisci M, Bolay H. Updated review on the link between cortical spreading depression and headache disorders. Expert Rev Neurother 2021; 21:1069-1084. [PMID: 34162288 DOI: 10.1080/14737175.2021.1947797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Experimental animal studies have revealed mechanisms that link cortical spreading depression (CSD) to the trigeminal activation mediating lateralized headache. However, conventional CSD as seen in lissencephalic brain is insufficient to explain some clinical features of aura and migraine headache. AREAS COVERED The importance of CSD in headache development including dysfunction of the thalamocortical network, neuroinflammation, calcitonin gene-related peptide, transgenic models, and the role of CSD in migraine triggers, treatment options, neuromodulation and future directions are reviewed. EXPERT OPINION The conventional understanding of CSD marching across the hemisphere is invalid in gyrencephalic brains. Thalamocortical dysfunction and interruption of functional cortical network systems by CSD, may provide alternative explanations for clinical manifestations of migraine phases including aura. Not all drugs showing CSD blocking properties in lissencephalic brains, have efficacy in migraine headache and monoclonal antibodies against CGRP ligand/receptors which are effective in migraine treatment, have no impact on aura in humans or CSD properties in rodents. Functional networks and molecular mechanisms mediating and amplifying the effects of limited CSD in migraine brain remain to be investigated to define new targets.
Collapse
Affiliation(s)
- Doga Vuralli
- Department of Neurology and Algology, Gazi University Faculty of Medicine, Besevler, Ankara, Turkey.,Neuropsychiatry Center, Gazi University, Besevler, Ankara, Turkey.,Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey
| | - Hulya Karatas
- Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey.,Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Muge Yemisci
- Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey.,Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Hayrunnisa Bolay
- Department of Neurology and Algology, Gazi University Faculty of Medicine, Besevler, Ankara, Turkey.,Neuropsychiatry Center, Gazi University, Besevler, Ankara, Turkey.,Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey
| |
Collapse
|
15
|
Crivellaro G, Tottene A, Vitale M, Melone M, Casari G, Conti F, Santello M, Pietrobon D. Specific activation of GluN1-N2B NMDA receptors underlies facilitation of cortical spreading depression in a genetic mouse model of migraine with reduced astrocytic glutamate clearance. Neurobiol Dis 2021; 156:105419. [PMID: 34111520 DOI: 10.1016/j.nbd.2021.105419] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/29/2021] [Accepted: 06/04/2021] [Indexed: 01/28/2023] Open
Abstract
Migraine is a common but poorly understood sensory circuit disorder. Mouse models of familial hemiplegic migraine (FHM, a rare monogenic form of migraine with aura) show increased susceptibility to cortical spreading depression (CSD, the phenomenon that underlies migraine aura and can activate migraine headache mechanisms), allowing an opportunity to investigate the mechanisms of CSD and migraine onset. In FHM type 2 (FHM2) knock-in mice with reduced expression of astrocytic Na+, K+-ATPases, the reduced rate of glutamate uptake into astrocytes can account for the facilitation of CSD initiation. Here, we investigated the underlying mechanisms and show that the reduced rate of glutamate clearance in FHM2 mice results in increased amplitude and slowing of rise time and decay of the NMDA receptor (NMDAR) excitatory postsynaptic current (EPSC) elicited in layer 2/3 pyramidal cells by stimulation of neuronal afferents in somatosensory cortex slices. The relative increase in NMDAR activation in FHM2 mice is activity-dependent, being larger after high-frequency compared to low-frequency afferent activity. Inhibition of GluN1-N2B NMDARs, which hardly affected the NMDAR EPSC in wild-type mice, rescued the increased and prolonged activation of NMDARs as well as the facilitation of CSD induction and propagation in FHM2 mice. Our data suggest that the enhanced susceptibility to CSD in FHM2 is mainly due to specific activation of extrasynaptic GluN1-N2B NMDARs and point to these receptors as possible therapeutic targets for prevention of CSD and migraine.
Collapse
Affiliation(s)
- Giovanna Crivellaro
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Angelita Tottene
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Marina Vitale
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Marcello Melone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Italy Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy
| | - Giorgio Casari
- Vita Salute San Raffaele University and San Raffaele Scientific Institute, Milano, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Italy Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy; Fondazione di Medicina Molecolare, Università Politecnica delle Marche, Ancona, Italy
| | - Mirko Santello
- Institute of Pharmacology and Toxicology and Neuroscience Center Zurich, University of Zurich, CH-8057 Zurich, Switzerland
| | - Daniela Pietrobon
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Padova Neuroscience Center, University of Padova, CNR Institute of Neuroscience, 35131 Padova, Italy.
| |
Collapse
|
16
|
Just N. Proton functional magnetic resonance spectroscopy in rodents. NMR IN BIOMEDICINE 2021; 34:e4254. [PMID: 31967711 DOI: 10.1002/nbm.4254] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/04/2019] [Accepted: 12/16/2019] [Indexed: 06/10/2023]
Abstract
Proton functional magnetic resonance spectroscopy (1 H-fMRS) in the human brain is able to assess and quantify the metabolic response due to localized brain activity. Currently, 1 H-fMRS of the human brain is complementary to functional magnetic resonance imaging (fMRI) and a recommended technique at high field strengths (>7 T) for the investigation of neurometabolic couplings, thereby providing insight into the mechanisms underlying brain activity and brain connectivity. Understanding typical healthy brain metabolism during a task is expected to provide a baseline from which to detect and characterize neurochemical alterations associated with various neurological or psychiatric disorders and diseases. It is of paramount importance to resolve fundamental questions related to the regulation of neurometabolic processes. New techniques such as optogenetics may be coupled to fMRI and fMRS to bring more specificity to investigations of brain cell populations during cerebral activation thus enabling a higher link to molecular changes and therapeutic advances. These rather novel techniques are mainly available for rodent applications and trigger renewed interest in animal fMRS. However, rodent fMRS remains fairly confidential due to its inherent low signal-to-noise ratio and its dependence on anesthesia. For instance, the accurate determination of metabolic concentration changes during stimulation requires robust knowledge of the physiological environment of the measured region of interest linked to anesthesia in most cases. These factors may also have a strong influence on B0 homogeneity. Therefore, a degree of calibration of the stimulus strength and duration may be needed for increased knowledge of the underpinnings of cerebral activity. Here, we propose an early review of the current status of 1 H-fMRS in rodents and summarize current difficulties and future perspectives.
Collapse
Affiliation(s)
- Nathalie Just
- Department of Clinical Radiology, University Hospital Münster, Germany
- INRAE, Centre, Tours Val de Loire, France
| |
Collapse
|
17
|
Hyperhomocysteinemia increases susceptibility to cortical spreading depression associated with photophobia, mechanical allodynia, and anxiety in rats. Behav Brain Res 2021; 409:113324. [PMID: 33915239 DOI: 10.1016/j.bbr.2021.113324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 11/23/2022]
Abstract
Epidemiological data suggest that elevated homocysteine is associated with migraine with aura. However, how homocysteine contributes to migraine is still unclear. Here, we tested whether hyperhomocysteinemia (hHCY) promotes cortical spreading depression (CSD), a phenomenon underlying migraine with aura, and whether hHCY contributes to pain behavior. hHCY was induced by dietary methionine in female rats while the testing was performed on their 6-8week-old offspring. CSD and multiple unit activity (MUA) induced by KCl were recorded from the primary somatosensory cortex, S1, using multichannel electrodes. In hHCY rats, compared to control, we found: i) higher probability of CSD occurrence; ii) induction of CSD by lower concentrations of KCl; iii) faster horizontal propagation of CSD; iv) smaller CSD with longer duration; v) higher frequency of MUA at CSD onset along with slower reappearance. Rats with hHCY demonstrated high level of locomotor activity and grooming while spent less time in the central area of the open field, indicating anxiety. These animals showed light sensitivity and facial mechanical allodinia. Thus, hHCY acquired at birth promotes multiple features of migraine such as higher cortical excitability, mechanical allodynia, photophobia, and anxiety. Our results provide the first experimental explanation for the higher occurrence of migraine with aura in patients with hHCY.
Collapse
|
18
|
Sarawagi A, Soni ND, Patel AB. Glutamate and GABA Homeostasis and Neurometabolism in Major Depressive Disorder. Front Psychiatry 2021; 12:637863. [PMID: 33986699 PMCID: PMC8110820 DOI: 10.3389/fpsyt.2021.637863] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
Major depressive disorder (MDD) is a leading cause of distress, disability, and suicides. As per the latest WHO report, MDD affects more than 260 million people worldwide. Despite decades of research, the underlying etiology of depression is not fully understood. Glutamate and γ-aminobutyric acid (GABA) are the major excitatory and inhibitory neurotransmitters, respectively, in the matured central nervous system. Imbalance in the levels of these neurotransmitters has been implicated in different neurological and psychiatric disorders including MDD. 1H nuclear magnetic resonance (NMR) spectroscopy is a powerful non-invasive method to study neurometabolites homeostasis in vivo. Additionally, 13C-NMR spectroscopy together with an intravenous administration of non-radioactive 13C-labeled glucose or acetate provides a measure of neural functions. In this review, we provide an overview of NMR-based measurements of glutamate and GABA homeostasis, neurometabolic activity, and neurotransmitter cycling in MDD. Finally, we highlight the impact of recent advancements in treatment strategies against a depressive disorder that target glutamate and GABA pathways in the brain.
Collapse
Affiliation(s)
- Ajay Sarawagi
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Narayan Datt Soni
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Anant Bahadur Patel
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
19
|
Shatillo A, Lipponen A, Salo RA, Tanila H, Verkhratsky A, Giniatullin R, Gröhn OH. Spontaneous BOLD waves - A novel hemodynamic activity in Sprague-Dawley rat brain detected by functional magnetic resonance imaging. J Cereb Blood Flow Metab 2019; 39:1949-1960. [PMID: 29690796 PMCID: PMC6775581 DOI: 10.1177/0271678x18772994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We report spontaneous hemodynamic activity termed "Spontaneous BOLD Waves" (SBWs) detected by BOLD fMRI in Sprague-Dawley rats under medetomidine anesthesia. These SBWs, which lasted several minutes, were observed in cortex, thalamus and hippocampus. The SBWs' correlates were undetectable in electrophysiological recordings, suggesting an exclusive gliovascular phenomenon dissociated from neuronal activity. SBWs were insensitive to the NMDA receptors antagonist MK-801 but were inhibited by the α1-adrenoceptor blocker prazosin. Since medetomidine is a potent agonist of α2 adrenoceptors, we suggested that imbalance in α1/α2 receptor-mediated signalling pathways alter the vascular reactivity leading to SBWs. The frequency of SBWs increased with intensity of mechanical lung ventilation despite the stable pH levels. In summary, we present a novel type of propagating vascular brain activity without easily detectable underlying neuronal activity, which can be utilized to study the mechanisms of vascular reactivity in functional and pharmacological MRI and has practical implications for designing fMRI experiments in anesthetized animals.
Collapse
Affiliation(s)
- Artem Shatillo
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Arto Lipponen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Raimo A Salo
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Heikki Tanila
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Rashid Giniatullin
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Lab of Neurobiology, Kazan Federal University, Kazan, Russia
| | - Olli H Gröhn
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
20
|
Long lasting behavioral and electrophysiological action of early administration of guanosine: Analysis in the adult rat brain. Brain Res Bull 2019; 150:266-271. [PMID: 31181322 DOI: 10.1016/j.brainresbull.2019.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/23/2019] [Accepted: 06/04/2019] [Indexed: 01/01/2023]
Abstract
Guanosine (GUO) is a guanine-based purine that has been extensively described in the literature as an endogenous nucleoside with participation in brain cell signalling pathways. Here, we evaluated whether chronic treatment with exogenous guanosine during brain development altered behavioral and electrophysiological parameters in adulthood. Rat pups received a daily intraperitoneal injection of 10, 50 or 100 mg/ kg/day GUO, or saline solution or no treatment (naive group) from postnatal (P) day 7 to P27. At P 60-65 the animals were behaviorally tested in the Elevated Plus-Maze (EPM). On P90-100, the electrophysiological phenomenon known as cortical spreading depression (CSD) was recorded on the right cortical surface for 4 h. With the EPM task, GUO treatment was associated with a significant increase in rearing behavior and a non-significant trend towards anxiogenic behavior. In a dose-dependent manner, GUO significantly (p < 0.01) increased weight gain on P90, and reduced the CSD propagation velocity from 3.42 ± 0.10 and 3.43 ± 0.10 mm/min in the Naive and Vehicle controls, respectively, to 3.05 ± 0.12 mm/min, 2.87 ± 0.07 mm/min and 2.25 ± 0.25 mm/min in the groups treated with 10, 50 and 100 mg/kg/d GUO, respectively. The results confirmed the hypothesis that the chronic treatment with GUO early in life modulates CSD and body weight. Data on CSD propagation suggest that, besides its suppressing action on glutamatergic transmission (via enhancement of astrocytic glutamate uptake), GUO might act as an antioxidant in the brain, a hypothesis that deserves further exploration.
Collapse
|
21
|
Xiao T, Wang Y, Wei H, Yu P, Jiang Y, Mao L. Electrochemical Monitoring of Propagative Fluctuation of Ascorbate in the Live Rat Brain during Spreading Depolarization. Angew Chem Int Ed Engl 2019; 58:6616-6619. [DOI: 10.1002/anie.201901035] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/20/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Tongfang Xiao
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
| | - Yuexiang Wang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
| | - Huan Wei
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Ping Yu
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Ying Jiang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
| | - Lanqun Mao
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
22
|
Xiao T, Wang Y, Wei H, Yu P, Jiang Y, Mao L. Electrochemical Monitoring of Propagative Fluctuation of Ascorbate in the Live Rat Brain during Spreading Depolarization. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201901035] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Tongfang Xiao
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
| | - Yuexiang Wang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
| | - Huan Wei
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Ping Yu
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Ying Jiang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
| | - Lanqun Mao
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Analytical Chemistry for Living BiosystemsInstitute of Chemistry, theChinese Academy of Sciences (CAS)CAS Research/Education Center for Excellence in Molecular Science Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
23
|
Accioly NE, Guedes RCA. Neonatal treatment with ovarian hormones and suckling among distinct litter sizes: Differential effects on recognition memory and spreading depression at adulthood. Nutr Neurosci 2019; 22:174-184. [PMID: 28891432 DOI: 10.1080/1028415x.2017.1358472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Ovarian hormones (OH) and early malnutrition may affect the developing brain, with repercussions on behavioral and excitability-dependent processes. However, the possible synergistic effects of both factors have not been analyzed. In this study, we investigated the effect of treatment in early life with OH and suckling among distinct litter sizes on recognition memory, anxiety behavior, and the excitability-dependent phenomenon known as cortical spreading depression (CSD). METHODS Female Wistar rats were suckled under favorable and unfavorable lactation, corresponding to litters with 9 and 15 pups (L9 and L15 groups, respectively). From postnatal days (P) 7 to 21, the animals received 50 µg/kg of β-estradiol or progesterone. From P80 to P84, we tested behavioral reactions. From P90 to P120, we analyzed CSD parameters. RESULTS Compared with the corresponding L9 groups, the OH-treated L15 groups performed worse in recognition memory tasks. No intergroup difference in the anxiety parameters was observed. Compared with naive and vehicle-treated controls, OH-treated groups displayed higher CSD velocities and amplitudes and shorter CSD durations. DISCUSSION Early treatment with OH facilitates recognition memory and CSD, and in association with unfavorable lactation (L15) impaired recognition memory, but not anxiety behavior, in the adult brain. OH treatment and L15 lactation condition seem to interact regarding OH action on memory, but not on CSD. Data suggest a long-lasting differential effect that might be related to the lasting hormonal action on brain excitability. We postulate and discuss the possibility that these findings may be implicated in human neurological diseases.
Collapse
|
24
|
de Iure A, Napolitano F, Beck G, Quiroga Varela A, Durante V, Sciaccaluga M, Mazzocchetti P, Megaro A, Tantucci M, Cardinale A, Punzo D, Mancini A, Costa C, Ghiglieri V, Tozzi A, Picconi B, Papa SM, Usiello A, Calabresi P. Striatal spreading depolarization: Possible implication in levodopa-induced dyskinetic-like behavior. Mov Disord 2019; 34:832-844. [PMID: 30759320 DOI: 10.1002/mds.27632] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/07/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Spreading depolarization (SD) is a transient self-propagating wave of neuronal and glial depolarization coupled with large membrane ionic changes and a subsequent depression of neuronal activity. Spreading depolarization in the cortex is implicated in migraine, stroke, and epilepsy. Conversely, spreading depolarization in the striatum, a brain structure deeply involved in motor control and in Parkinson's disease (PD) pathophysiology, has been poorly investigated. METHODS We characterized the participation of glutamatergic and dopaminergic transmission in the induction of striatal spreading depolarization by using a novel approach combining optical imaging, measurements of endogenous DA levels, and pharmacological and molecular analyses. RESULTS We found that striatal spreading depolarization requires the concomitant activation of D1-like DA and N-methyl-d-aspartate receptors, and it is reduced in experimental PD. Chronic l-dopa treatment, inducing dyskinesia in the parkinsonian condition, increases the occurrence and speed of propagation of striatal spreading depolarization, which has a direct impact on one of the signaling pathways downstream from the activation of D1 receptors. CONCLUSION Striatal spreading depolarization might contribute to abnormal basal ganglia activity in the dyskinetic condition and represents a possible therapeutic target. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Antonio de Iure
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy.,Laboratory of Experimental Neurophysiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy
| | - Francesco Napolitano
- Ceinge Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II,", Naples, Italy
| | - Goichi Beck
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ana Quiroga Varela
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Valentina Durante
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Miriam Sciaccaluga
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Petra Mazzocchetti
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Alfredo Megaro
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Michela Tantucci
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Antonella Cardinale
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy
| | - Daniela Punzo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples, Caserta, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation SDN, Via Gianturco, Naples, Italy
| | - Andrea Mancini
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Cinzia Costa
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
| | - Veronica Ghiglieri
- Laboratory of Neurophysiology, Santa Lucia Foundation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.,Department of Philosophy, Human, Social and Educational Sciences, University of Perugia, Perugia, Italy
| | - Alessandro Tozzi
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, Santa Lucia Foundation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Barbara Picconi
- Laboratory of Experimental Neurophysiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alessandro Usiello
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples, Caserta, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation SDN, Via Gianturco, Naples, Italy
| | - Paolo Calabresi
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, Santa Lucia Foundation, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
25
|
Ferulic Acid Improves Depressive-Like Behavior in Prenatally-Stressed Offspring Rats via Anti-Inflammatory Activity and HPA Axis. Int J Mol Sci 2019; 20:ijms20030493. [PMID: 30678337 PMCID: PMC6387299 DOI: 10.3390/ijms20030493] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 01/23/2023] Open
Abstract
Prenatal stress (PS) can increase the risk of nervous, endocrine and metabolic diseases, and immune dysfunction. Ferulic acid (FA) is a dietary phenolic acid that has pharmacological properties, including potent anti-inflammatory action. We used male, prenatally-stressed offspring rats to investigate the anti-depressive-like effects and possible anti-inflammatory mechanism of FA. We determined the animal behaviors, and the mRNA expression and concentration of inflammatory cytokines, and HPA axis. In addition, we assessed the modulation of hippocampal nuclear factor-κB (NF-κB) activation, neuronal nitric oxide synthase (nNOS) and glucocorticoid receptors (GR) expression via western blotting and immunohistochemistry. Administration of FA (12.5, 25, and 50 mg/kg/day, i.g.) for 28 days markedly increased sucrose intake, and decreased immobility time and total number of crossings, center crossings, rearing, and grooming in the male PS offspring. FA significantly reduced IL-6, IL-1β, and TNF-α concentration and increased IL-10 concentration in male, prenatally-stressed offspring, stimulated by the NF-κB pathway. In addition, FA inhibited interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α), and increased interleukin-10 (IL-10) mRNA and protein expression. Furthermore, FA markedly decreased the serum adrenocorticotropin (ACTH) and corticosterone concentration by the increase of GR protein expression. Taken together, this study revealed that FA has anti-depressive-like effects in male, prenatally-stressed offspring, partially due to its anti-inflammatory activity and hypothalamic-pituitary-adrenal (HPA) axis.
Collapse
|
26
|
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: 3.2] [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
|
27
|
Klass A, Sánchez-Porras R, Santos E. Systematic review of the pharmacological agents that have been tested against spreading depolarizations. J Cereb Blood Flow Metab 2018; 38:1149-1179. [PMID: 29673289 PMCID: PMC6434447 DOI: 10.1177/0271678x18771440] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Spreading depolarization (SD) occurs alongside brain injuries and it can lead to neuronal damage. Therefore, pharmacological modulation of SD can constitute a therapeutic approach to reduce its detrimental effects and to improve the clinical outcome of patients. The major objective of this article was to produce a systematic review of all the drugs that have been tested against SD. Of the substances that have been examined, most have been shown to modulate certain SD characteristics. Only a few have succeeded in significantly inhibiting SD. We present a variety of strategies that have been proposed to overcome the notorious harmfulness and pharmacoresistance of SD. Information on clinically used anesthetic, sedative, hypnotic agents, anti-migraine drugs, anticonvulsants and various other substances have been compiled and reviewed with respect to the efficacy against SD, in order to answer the question of whether a drug at safe doses could be of therapeutic use against SD in humans.
Collapse
Affiliation(s)
- Anna Klass
- Neurosurgery Department, University of Heidelberg, Heidelberg, Germany
| | | | - Edgar Santos
- Neurosurgery Department, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
28
|
E Silva-Gondim MB, de Souza TKM, Rodrigues MCA, Guedes RCA. Suckling in litters with different sizes, and early and late swimming exercise differentially modulates anxiety-like behavior, memory and electrocorticogram potentiation after spreading depression in rats. Nutr Neurosci 2017; 22:464-473. [PMID: 29183255 DOI: 10.1080/1028415x.2017.1407472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Analyze the hypothesis that swimming exercise, in rats suckled under distinct litter sizes, alters behavioral parameters suggestive of anxiety and recognition memory, and the electrocorticogram potentiation that occurs after the excitability-related phenomenon that is known as cortical spreading depression (CSD). METHODS Male Wistar rats were suckled in litters with six or 12 pups (L6 and L12 groups). Animals swam at postnatal days (P) 8-23, or P60-P75 (early-exercised or late-exercised groups, respectively), or remained no-exercised. Behavioral tests (open field - OF and object recognition - OR) were conducted between P77 and P80. Between P90 and P120, ECoG was recorded for 2 hours. After this 'baseline' recording, CSD was elicited every 30 minutes over the course of 2 hours. RESULTS Early swimming enhanced the number of entries and the percentage of time in the OF-center (P < 0.05). In animals that swam later, this effect occurred in the L6 group only. Compared to the corresponding sedentary groups, OR-test showed a better memory in the L6 early exercised rats, and a worse memory in all other groups (P < 0.05). In comparison to baseline values, ECoG amplitudes after CSD increased 14-43% for all groups (P < 0.05). In the L6 condition, early swimming and late swimming, respectively, reduced and enhanced the magnitude of the post-CSD ECoG potentiation in comparison with the corresponding L6 no-exercised groups (P < 0.05). DISCUSSION Our data suggest a differential effect of early- and late-exercise on the behavioral and electrophysiological parameters, suggesting an interaction between the age of exercise and the nutritional status during lactation.
Collapse
Affiliation(s)
- Mariana Barros E Silva-Gondim
- a Laboratory of Nutrition Physiology, Departamento de Nutrição , Universidade Federal de Pernambuco , CEP 50670-901 Recife , PE , Brazil
| | - Thays Kallyne Marinho de Souza
- a Laboratory of Nutrition Physiology, Departamento de Nutrição , Universidade Federal de Pernambuco , CEP 50670-901 Recife , PE , Brazil.,b Colegiado de Nutrição , Universidade de Pernambuco , CEP 56328-903 Petrolina , Pernambuco , Brazil
| | | | - Rubem Carlos Araújo Guedes
- a Laboratory of Nutrition Physiology, Departamento de Nutrição , Universidade Federal de Pernambuco , CEP 50670-901 Recife , PE , Brazil
| |
Collapse
|
29
|
Vitor-de-Lima SM, Medeiros LDB, Benevides RDDL, Dos Santos CN, Lima da Silva NO, Guedes RCA. Monosodium glutamate and treadmill exercise: Anxiety-like behavior and spreading depression features in young adult rats. Nutr Neurosci 2017; 22:435-443. [PMID: 29125056 DOI: 10.1080/1028415x.2017.1398301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES The route of administration is an important factor in determining the action of some drugs. We previously demonstrated that subcutaneous monosodium glutamate (MSG) accelerated cortical spreading depression (CSD) in the rat and that treadmill exercise attenuated this effect. This study evaluated whether other routes of administration exert the same action by testing orogastric (gavage) and topical cortical MSG administration in treadmill-exercised and sedentary rats. Additionally, in the orogastric treatment we tested anxiety-like behavior. METHODS Exercised and sedentary rats received per gavage water or MSG (1 or 2 g/kg) daily from postnatal (P) day 7 to 27. Behavioral tests (open field and elevated plus-maze) occurred at P53 ± 3. At P56 ± 3, we analyzed CSD parameters (velocity, amplitude, and duration of the negative potential change). Other three groups of rats received an MSG solution (25, 50 or 75 mg/ml) topically to the intact dura mater during CSD recording. RESULTS MSG-gavage increased anxiety-like behavior and the CSD velocities compared with water-treated controls (P < 0.05). Exercise decelerated CSD. In contrast to gavage, which accelerated CSD, topical MSG dose-dependently and reversibly impaired CSD propagation, reduced CSD amplitude and increased CSD duration (P < 0.05). CONCLUSIONS The exercise-dependent attenuation of the effects of MSG confirms our previous results in rats treated subcutaneously with MSG. CSD results suggest two distinct mechanisms for gavage and topical MSG administration. Additionally, data suggest that exercise can help protect the developing and adult brain against the deleterious actions of MSG.
Collapse
|
30
|
Ferrari A, Rustichelli C, Baraldi C. Glutamate receptor antagonists with the potential for migraine treatment. Expert Opin Investig Drugs 2017; 26:1321-1330. [PMID: 29050521 DOI: 10.1080/13543784.2017.1395411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Preclinical, clinical, and other (e.g., genetic) evidence support the concept that migraine susceptibility may at least partially result from a glutamatergic system disorder. Therefore, the receptors of the glutamatergic system are considered relatively new targets for investigational drugs to treat migraine. Investigational and established glutamate receptor antagonists (GluRAs) have been shown to possess antinociceptive properties in preclinical models of trigeminovascular nociception and have been evaluated in clinical trials. This review focuses on preclinical and clinical studies of GluRAs for the treatment of migraine. Areas covered: A PubMed database search (from 1987 to December 2016) and a review of published studies on GluRAs in migraine were conducted. Expert opinion: All published clinical trials of investigational GluRAs have been unsuccessful in establishing benefit for acute migraine treatment. Clinical trial results contrast with the preclinical data, suggesting that glutamate (Glu) does not play a decisive role after the attack has already been triggered. These antagonists may instead be useful for migraine prophylaxis. Improving patient care requires further investigating and critically analyzing the role of Glu in migraine, designing experimental models to study more receptors and their corresponding antagonists, and identifying biomarkers to facilitate trials designed to target specific subgroups of migraine patients.
Collapse
Affiliation(s)
- Anna Ferrari
- a Unit of Medical Toxicology, Headache and Drug Abuse Centre; Department of Diagnostic, Clinical and Public Health Medicine , University of Modena and Reggio Emilia , Modena , Italy
| | - Cecilia Rustichelli
- b Department of Life Sciences , University of Modena and Reggio Emilia , Modena , Italy
| | - Carlo Baraldi
- a Unit of Medical Toxicology, Headache and Drug Abuse Centre; Department of Diagnostic, Clinical and Public Health Medicine , University of Modena and Reggio Emilia , Modena , Italy
| |
Collapse
|
31
|
Kurauchi Y, Mokudai K, Mori A, Sakamoto K, Nakahara T, Morita M, Kamimura A, Ishii K. l-Citrulline ameliorates cerebral blood flow during cortical spreading depression in rats: Involvement of nitric oxide- and prostanoids-mediated pathway. J Pharmacol Sci 2017; 133:146-155. [PMID: 28325558 DOI: 10.1016/j.jphs.2017.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 12/21/2022] Open
Abstract
l-Citrulline is a potent precursor of l-arginine, and exerts beneficial effect on cardiovascular system via nitric oxide (NO) production. Migraine is one of the most popular neurovascular disorder, and imbalance of cerebral blood flow (CBF) observed in cortical spreading depression (CSD) contributes to the mechanism of migraine aura. Here, we investigated the effect of l-citrulline on cardiovascular changes to KCl-induced CSD. in rats. Intravenous injection of l-citrulline prevented the decrease in CBF, monitored by laser Doppler flowmetry, without affecting mean arterial pressure and heart rate during CSD. Moreover, l-citrulline attenuated propagation velocity of CSD induced by KCl. The effect of l-citrulline on CBF change was prevented by l-NAME, an inhibitor of NO synthase, but not by indomethacin, an inhibitor of cyclooxygenase. On the other hand, attenuation effect of l-citrulline on CSD propagation velocity was prevented not only by l-NAME but also by indomethacin. In addition, propagation velocity of CSD was attenuated by intravenous injection of NOR3, a NO donor, which was diminished by ODQ, an inhibitor of soluble guanylyl cyclase. These results suggest that NO/cyclic GMP- and prostanoids-mediated pathway differently contribute to the effect of l-citrulline on the maintenance of CBF.
Collapse
Affiliation(s)
- Yuki Kurauchi
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| | - Koichi Mokudai
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Masahiko Morita
- Healthcare Products Development Center, Kyowa Hakko Bio Co., Ltd., 2 Miyukigaoka, Tsukuba-shi, Ibaraki 305-0841, Japan
| | - Ayako Kamimura
- Healthcare Products Development Center, Kyowa Hakko Bio Co., Ltd., 2 Miyukigaoka, Tsukuba-shi, Ibaraki 305-0841, Japan
| | - Kunio Ishii
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
32
|
Lauritsen C, Mazuera S, Lipton RB, Ashina S. Intravenous ketamine for subacute treatment of refractory chronic migraine: a case series. J Headache Pain 2016; 17:106. [PMID: 27878523 PMCID: PMC5120050 DOI: 10.1186/s10194-016-0700-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/17/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Refractory migraine is a challenging condition with great impact on health related quality of life. Intravenous (IV) ketamine has been previously used to treat various refractory pain conditions. We present a series of patients with refractory migraine treated with intravenous ketamine in the hospital setting. METHODS Based on retrospective chart review, we identified six patients with refractory migraine admitted from 2010 through 2014 for treatment with intravenous ketamine. Ketamine was administered using a standard protocol starting with a dose of 0.1 mg/kg/hr and increased by 0.1 mg/kg/hr every 3 to 4 h as tolerated until the target pain score of 3/10 was achieved and maintained for at least 8 h. Visual Analogue Scale (VAS) scores at time of hospital admission were obtained as well as average baseline VAS scores prior to ketamine infusion. A phone interview was conducted for follow-up of migraine response in the 3 to 6 months following ketamine infusion. RESULTS The study sample had a median age of 36.5 years (range 29-54) and 83% were women. Pre-treatment pain scores ranged from 9 to 10. All patients achieved a target pain level of 3 or less for 8 h; the average ketamine infusion rate at target was 0.34 mg/kg/hour (range 0.12-0.42 mg/kg/hr). One patient reported a transient out-of-body hallucination following an increase in the infusion rate, which resolved after decreasing the rate. There were no other significant side effects. CONCLUSION IV ketamine was safely administered in the hospital setting to patients with refractory chronic migraine. Treatment was associated with short term improvement in pain severity in 6 of 6 patients with refractory chronic migraine. Prospective placebo-controlled trials are needed to assess short term and long-term efficacy of IV ketamine in refractory chronic migraine.
Collapse
Affiliation(s)
- Clinton Lauritsen
- Department of Neurology, Thomas Jefferson University, Jefferson Headache Center, Philadelphia, PA, USA.
| | - Santiago Mazuera
- Department of Neurology, Thomas Jefferson University, Jefferson Headache Center, Philadelphia, PA, USA
| | - Richard B Lipton
- Department of Neurology, Montefiore Headache Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sait Ashina
- Department of Neurology, New York University School of Medicine, NYU Langone Medical Center, NYU Lutheran Headache Center, New York, NY, USA
| |
Collapse
|
33
|
Abushik PA, Bart G, Korhonen P, Leinonen H, Giniatullina R, Sibarov DA, Levonen AL, Malm T, Antonov SM, Giniatullin R. Pro-nociceptive migraine mediator CGRP provides neuroprotection of sensory, cortical and cerebellar neurons via multi-kinase signaling. Cephalalgia 2016; 37:1373-1383. [PMID: 27884929 DOI: 10.1177/0333102416681588] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Blocking the pro-nociceptive action of CGRP is one of the most promising approaches for migraine prophylaxis. The aim of this study was to explore a role for CGRP as a neuroprotective agent for central and peripheral neurons. Methods The viability of isolated rat trigeminal, cortical and cerebellar neurons was tested by fluorescence vital assay. Engagement of Nrf2 target genes was analyzed by qPCR. The neuroprotective efficacy of CGRP in vivo was tested in mice using a permanent cerebral ischemia model. Results CGRP prevented apoptosis induced by the amino acid homocysteine in all three distinct neuronal populations. Using a set of specific kinase inhibitors, we show the role of multi-kinase signaling pathways involving PKA and CaMKII in neuronal survival. Forskolin triggered a very similar signaling cascade, suggesting that cAMP is the main upstream trigger for multi-kinase neuroprotection. The specific CGRP antagonist BIBN4096 reduced cellular viability, lending further support to the proposed neuroprotective function of CGRP. Importantly, CGRP was neuroprotective against permanent ischemia in mice. Conclusion Our data show an unexpected 'positive' role for the endogenous pro-nociceptive migraine mediator CGRP, suggesting more careful examination of migraine prophylaxis strategy based on CGRP antagonism although it should be noted that homocysteine induced apoptosis in primary neuronal cell culture might not necessarily reproduce all the features of cell loss in the living organism.
Collapse
Affiliation(s)
- Polina A Abushik
- 1 Department of Neurobiology, University of Eastern Finland, Kuopio, Finland.,2 Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Laboratory of Comparative Neurophysiology, Saint-Petersburg, Russia
| | - Geneviève Bart
- 1 Department of Neurobiology, University of Eastern Finland, Kuopio, Finland
| | - Paula Korhonen
- 1 Department of Neurobiology, University of Eastern Finland, Kuopio, Finland
| | - Hanna Leinonen
- 3 Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Raisa Giniatullina
- 1 Department of Neurobiology, University of Eastern Finland, Kuopio, Finland
| | - Dmitry A Sibarov
- 2 Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Laboratory of Comparative Neurophysiology, Saint-Petersburg, Russia
| | - Anna-Liisa Levonen
- 3 Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- 1 Department of Neurobiology, University of Eastern Finland, Kuopio, Finland
| | - Sergei M Antonov
- 2 Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Laboratory of Comparative Neurophysiology, Saint-Petersburg, Russia
| | - Rashid Giniatullin
- 1 Department of Neurobiology, University of Eastern Finland, Kuopio, Finland.,4 Laboratory of Neurobiology, Department of Physiology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
34
|
Neonatal l-glutamine modulates anxiety-like behavior, cortical spreading depression, and microglial immunoreactivity: analysis in developing rats suckled on normal size- and large size litters. Amino Acids 2016; 49:337-346. [DOI: 10.1007/s00726-016-2365-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/11/2016] [Indexed: 12/19/2022]
|
35
|
Gasparini CF, Smith RA, Griffiths LR. Genetic insights into migraine and glutamate: a protagonist driving the headache. J Neurol Sci 2016; 367:258-68. [PMID: 27423601 DOI: 10.1016/j.jns.2016.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 05/11/2016] [Accepted: 06/08/2016] [Indexed: 12/12/2022]
Abstract
Migraine is a complex polygenic disorder that continues to be a great source of morbidity in the developed world with a prevalence of 12% in the Caucasian population. Genetic and pharmacological studies have implicated the glutamate pathway in migraine pathophysiology. Glutamate profoundly impacts brain circuits that regulate core symptom domains in a range of neuropsychiatric conditions and thus remains a "hot" target for drug discovery. Glutamate has been implicated in cortical spreading depression (CSD), the phenomenon responsible for migraine with aura and in animal models carrying FHM mutations. Genotyping case-control studies have shown an association between glutamate receptor genes, namely, GRIA1 and GRIA3 with migraine with indirect supporting evidence from GWAS. New evidence localizes PRRT2 at glutamatergic synapses and shows it affects glutamate signalling and glutamate receptor activity via interactions with GRIA1. Glutamate-system defects have also been recently implicated in a novel FHM2 ATP1A2 disease-mutation mouse model. Adding to the growing evidence neurophysiological findings support a role for glutamate in cortical excitability. In addition to the existence of multiple genes to choreograph the functions of fast-signalling glutamatergic neurons, glutamate receptor diversity and regulation is further increased by the post-translational mechanisms of RNA editing and miRNAs. Ongoing genetic studies, GWAS and meta-analysis implicate neurogenic mechanisms in migraine pathology and the first genome-wide associated locus for migraine on chromosome X. Finally, in addition to glutamate modulating therapies, the kynurenine pathway has emerged as a candidate for involvement in migraine pathophysiology. In this review we discuss recent genetic evidence and glutamate modulating therapies that bear on the hypothesis that a glutamatergic mechanism may be involved in migraine susceptibility.
Collapse
Affiliation(s)
- Claudia F Gasparini
- Menzies Health Institute Queensland, Griffith University Gold Coast, Parklands Drive, Southport, QLD 4222, Australia
| | - Robert A Smith
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Musk Ave, Kelvin Grove, QLD 4059, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Musk Ave, Kelvin Grove, QLD 4059, Australia.
| |
Collapse
|
36
|
Bu F, Du R, Li Y, Quinn JP, Wang M. NR2A contributes to genesis and propagation of cortical spreading depression in rats. Sci Rep 2016; 6:23576. [PMID: 27001011 PMCID: PMC4802317 DOI: 10.1038/srep23576] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/08/2016] [Indexed: 11/09/2022] Open
Abstract
Cortical spreading depression (CSD) is a transient propagating excitation of synaptic activity followed by depression, which is implicated in migraine. Increasing evidence points to an essential role of NR2A-containing NMDA receptors in CSD propagation in vitro; however, whether these receptors mediate CSD genesis in vivo requires clarification and the role of NR2A on CSD propagation is still under debate. Using in vivo CSD in rats with electrophysiology and in vitro CSD in chick retina with intrinsic optical imaging, we addressed the role of NR2A in CSD. We demonstrated that NVP-AAM077, a potent antagonist for NR2A-containing receptors, perfused through microdialysis probes, markedly reduced cortex susceptibility to CSD, but also reduced magnitude of CSD genesis in rats. Additionally, NVP-AAM077 at 0.3 nmol perfused into the contralateral ventricle, considerably suppressed the magnitude of CSD propagation wave and propagation rate in rats. This reduction in CSD propagation was also observed with TCN-201, a negative allosteric modulator selective for NR2A, at 3 μM, in the chick retina. Our data provides strong evidence that NR2A subunit contributes to CSD genesis and propagation, suggesting drugs selectively antagonizing NR2A-containing receptors might constitute a highly specific strategy treating CSD associated migraine with a likely better safety profile.
Collapse
Affiliation(s)
- Fan Bu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China.,Centre for Neuroscience, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| | - Ruoxing Du
- Centre for Neuroscience, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| | - Yi Li
- Department of Applied Chemistry, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| | - John P Quinn
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Minyan Wang
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China.,Centre for Neuroscience, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| |
Collapse
|
37
|
Jia Y, Zhou J, Bu F, Wang M. Synergistic Suppression of Cortical Spreading Depression under NR2A and NR2B Inhibition. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/pp.2015.612059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|