Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 567
Filter
1.
Biol Res ; 57(1): 39, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867288

ABSTRACT

BACKGROUND: Spreading depression (SD) is an intriguing phenomenon characterized by massive slow brain depolarizations that affect neurons and glial cells. This phenomenon is repetitive and produces a metabolic overload that increases secondary damage. However, the mechanisms associated with the initiation and propagation of SD are unknown. Multiple lines of evidence indicate that persistent and uncontrolled opening of hemichannels could participate in the pathogenesis and progression of several neurological disorders including acute brain injuries. Here, we explored the contribution of astroglial hemichannels composed of connexin-43 (Cx43) or pannexin-1 (Panx1) to SD evoked by high-K+ stimulation in brain slices. RESULTS: Focal high-K+ stimulation rapidly evoked a wave of SD linked to increased activity of the Cx43 and Panx1 hemichannels in the brain cortex, as measured by light transmittance and dye uptake analysis, respectively. The activation of these channels occurs mainly in astrocytes but also in neurons. More importantly, the inhibition of both the Cx43 and Panx1 hemichannels completely prevented high K+-induced SD in the brain cortex. Electrophysiological recordings also revealed that Cx43 and Panx1 hemichannels critically contribute to the SD-induced decrease in synaptic transmission in the brain cortex and hippocampus. CONCLUSIONS: Targeting Cx43 and Panx1 hemichannels could serve as a new therapeutic strategy to prevent the initiation and propagation of SD in several acute brain injuries.


Subject(s)
Astrocytes , Connexin 43 , Connexins , Cortical Spreading Depression , Synaptic Transmission , Animals , Astrocytes/physiology , Connexins/metabolism , Cortical Spreading Depression/physiology , Cortical Spreading Depression/drug effects , Synaptic Transmission/physiology , Synaptic Transmission/drug effects , Connexin 43/metabolism , Male , Nerve Tissue Proteins/metabolism , Cerebral Cortex , Neurons/physiology , Hippocampus , Rats, Sprague-Dawley , Rats , Potassium/metabolism
2.
Eur J Pharmacol ; 977: 176718, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38849040

ABSTRACT

Nimodipine is used to prevent delayed ischemic deficit in patients with aneurysmal subarachnoid hemorrhage (aSAH). Spreading depolarization (SD) is recognized as a factor in the pathomechanism of aSAH and other acute brain injuries. Although nimodipine is primarily known as a cerebral vasodilator, it may have a more complex mechanism of action due to the expression of its target, the L-type voltage-gated calcium channels (LVGCCs) in various cells in neural tissue. This study was designed to investigate the direct effect of nimodipine on SD, ischemic tissue injury, and neuroinflammation. SD in control or nimodipine-treated live mouse brain slices was induced under physiological conditions using electrical stimulation, or by subjecting the slices to hypo-osmotic stress or mild oxygen-glucose deprivation (mOGD). SD was recorded applying local field potential recording or intrinsic optical signal imaging. Histological analysis was used to estimate tissue injury, the number of reactive astrocytes, and the degree of microglia activation. Nimodipine did not prevent SD occurrence in mOGD, but it did reduce the rate of SD propagation and the cortical area affected by SD. In contrast, nimodipine blocked SD occurrence in hypo-osmotic stress, but had no effect on SD propagation. Furthermore, nimodipine prevented ischemic injury associated with SD in mOGD. Nimodipine also exhibited anti-inflammatory effects in mOGD by reducing reactive astrogliosis and microglial activation. The results demonstrate that nimodipine directly inhibits SD, independent of nimodipine's vascular effects. Therefore, the use of nimodipine may be extended to treat acute brain injuries where SD plays a central role in injury progression.


Subject(s)
Brain Ischemia , Brain , Cortical Spreading Depression , Nimodipine , Animals , Nimodipine/pharmacology , Mice , Cortical Spreading Depression/drug effects , Male , Brain/drug effects , Brain/pathology , Brain/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Mice, Inbred C57BL , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Osmotic Pressure/drug effects
3.
Sci Rep ; 14(1): 10186, 2024 05 03.
Article in English | MEDLINE | ID: mdl-38702377

ABSTRACT

Spreading depolarizations (SDs) occur frequently in patients with malignant hemispheric stroke. In animal-based experiments, SDs have been shown to cause secondary neuronal damage and infarct expansion during the initial period of infarct progression. In contrast, the influence of SDs during the delayed period is not well characterized yet. Here, we analyzed the impact of SDs in the delayed phase after cerebral ischemia and the potential protective effect of ketamine. Focal ischemia was induced by distal occlusion of the left middle cerebral artery in C57BL6/J mice. 24 h after occlusion, SDs were measured using electrocorticography and laser-speckle imaging in three different study groups: control group without SD induction, SD induction with potassium chloride, and SD induction with potassium chloride and ketamine administration. Infarct progression was evaluated by sequential MRI scans. 24 h after occlusion, we observed spontaneous SDs with a rate of 0.33 SDs/hour which increased during potassium chloride application (3.37 SDs/hour). The analysis of the neurovascular coupling revealed prolonged hypoemic and hyperemic responses in this group. Stroke volume increased even 24 h after stroke onset in the SD-group. Ketamine treatment caused a lesser pronounced hypoemic response and prevented infarct growth in the delayed phase after experimental ischemia. Induction of SDs with potassium chloride was significantly associated with stroke progression even 24 h after stroke onset. Therefore, SD might be a significant contributor to delayed stroke progression. Ketamine might be a possible drug to prevent SD-induced delayed stroke progression.


Subject(s)
Brain Ischemia , Disease Progression , Ketamine , Mice, Inbred C57BL , Ketamine/pharmacology , Animals , Mice , Male , Brain Ischemia/prevention & control , Brain Ischemia/diagnostic imaging , Brain Ischemia/drug therapy , Disease Models, Animal , Magnetic Resonance Imaging , Cortical Spreading Depression/drug effects , Infarction, Middle Cerebral Artery
4.
Neurosci Lett ; 832: 137814, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38723760

ABSTRACT

Galanin (Gal) is a neuropeptide with the potential to ameliorate cortical spreading depolarization (CSD), an electrophysiological phenomenon occurring after brain injury or in migraine aura. Gal is expressed in all cortical neurons both in rat and in mouse cortices. Here we investigated whether the effect of Gal on CSD previously described in the rat is conserved in the mouse cortex. In rats, the topical application of Gal to the cortex for 1 h did not induce any change in CSD amplitudes, propagation velocity, or threshold of elicitation. Rather, topical application of Gal for 3 h was necessary to obtain a significant decrease in these CSD parameters and to develop a remarkable increase in the KCl threshold to elicit a CSD in rat cortex. In contrast, the topical application of Gal on cortical surface for 1 h in mice was sufficient to significantly attenuate CSD amplitudes and increase threshold. A thinner cortex, a faster diffusion or different affinity/expression of receptors for Gal are possible reasons to explain this difference in the time course between rats and mice. Our data are relevant to postulate Gal as a potential target for inhibition of CSD under pathological situations such as stroke or ischemia. SIGNIFICANCE STATEMENT: The neuropeptide Galanin (Gal) is expressed in all neurons throughout the cerebral cortex, both in rats and mice, and is able to reduce or even inhibit Cortical Spreading Depolarization, thus, Gal has the potential to control neuronal excitability that may identify Gal as a target in drug development against CSD.


Subject(s)
Cerebral Cortex , Cortical Spreading Depression , Galanin , Animals , Galanin/pharmacology , Galanin/metabolism , Cortical Spreading Depression/drug effects , Cortical Spreading Depression/physiology , Male , Mice , Rats , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Rats, Wistar
5.
J Cereb Blood Flow Metab ; 44(6): 1000-1012, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38140913

ABSTRACT

Cortical spreading depolarization (SD) imposes a massive increase in energy demand and therefore evolves as a target for treatment following acute brain injuries. Anesthetics are empirically used to reduce energy metabolism in critical brain conditions, yet their effect on metabolism during SD remains largely unknown. We investigated oxidative metabolism during SD in brain slices from Wistar rats. Extracellular potassium ([K+]o), local field potential and partial tissue oxygen pressure (ptiO2) were measured simultaneously. The cerebral metabolic rate of oxygen (CMRO2) was calculated using a reaction-diffusion model. By that, we tested the effect of clinically relevant concentrations of isoflurane on CMRO2 during SD and modeled tissue oxygenation for different capillary pO2 values. During SD, CMRO2 increased 2.7-fold, resulting in transient hypoxia in the slice core. Isoflurane decreased CMRO2, reduced peak [K+]o, and prolonged [K+]o clearance, which indicates reduced synaptic transmission and sodium-potassium ATPase inhibition. Modeling tissue oxygenation during SD illustrates the need for increased capillary pO2 levels to prevent hypoxia. In the absence thereof, isoflurane could improve tissue oxygenation by lowering CMRO2. Therefore, isoflurane is a promising candidate for pre-clinical studies on neuronal survival in conditions involving SD.


Subject(s)
Cortical Spreading Depression , Isoflurane , Oxygen , Rats, Wistar , Animals , Isoflurane/pharmacology , Cortical Spreading Depression/drug effects , Cortical Spreading Depression/physiology , Rats , Oxygen/metabolism , Anesthetics, Inhalation/pharmacology , Male , Hypoxia/metabolism , Potassium/metabolism , Oxygen Consumption/drug effects , Brain/metabolism , Brain/drug effects , Hypoxia, Brain/metabolism , Hypoxia, Brain/drug therapy
6.
J Neurophysiol ; 126(6): 1934-1942, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34731067

ABSTRACT

Spreading depolarization (SD) is a slowly propagating wave of neuronal and glial depolarization. A growing number of studies show that SD and SD-like phenomena play a role in neurological disorders such as migraine, stroke, and traumatic brain injury. Despite the clinical importance of SD, its underlying molecular and cellular mechanisms remain elusive, possibly because of insufficient animal model allowing genetic manipulation. Such a model would also allow high-throughput screening for SD-suppressing drug development. To address this, we developed a novel experimental system to study SD using zebrafish. Electrophysiological recordings in the immobilized adult zebrafish revealed that increasing extracellular potassium concentration elicited SD with a large and long-lasting negative shift of direct current (DC) potential in the optic tectum. It also reduced the oscillatory activity in the extracellular field potential and increased the expression of the immediate early gene c-fos. Pharmacological blocking of the N-methyl-d-aspartate (NMDA) glutamate receptor attenuated the propagation of SD, suggesting that glutamatergic neurotransmission mediated tectal SD in zebrafish. Our analyses revealed that the zebrafish tectum and rodent cortex had similar SD kinetics. The current study provides electrophysiological and pharmacological evidence that zebrafish SD and mammal SD are comparable. This zebrafish SD model is suitable for genetic manipulation and cost-effective high-throughput screening. It could pave the way to novel diagnostic and therapeutic methods applicable to SD-associated neurological disorders.NEW & NOTEWORTHY Previous studies have implicated spreading depolarization (SD) in stroke and migraine. Here, we demonstrate SD, for the first time, in the adult zebrafish tectum showing waveform kinetics, c-fos expression, and attenuation by N-methyl-d-aspartate glutamate receptor blocker as observed in the rodent cortex. Since the zebrafish is an animal model amenable to genetic manipulation and chemical screening, this result could pave the way to novel diagnostic and therapeutic methods applicable to SD-associated neurological disorders.


Subject(s)
Cerebral Cortex , Cortical Spreading Depression , Excitatory Amino Acid Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Superior Colliculi , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Cortical Spreading Depression/drug effects , Cortical Spreading Depression/physiology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Superior Colliculi/drug effects , Superior Colliculi/physiology , Zebrafish
7.
World Neurosurg ; 155: e704-e715, 2021 11.
Article in English | MEDLINE | ID: mdl-34500101

ABSTRACT

OBJECTIVE: Cortical spreading depolarization (CSD), cortical spreading ischemia (CSI), and early brain injury are involved in the occurrence of delayed brain ischemia after subarachnoid hemorrhage (SAH). We tested whether local application of magnesium (Mg) sulfate solution suppressed CSD and CSI, and decreased brain damage in a rat SAH-mimicking model. METHODS: Nitric oxide synthase inhibitor L-NG-nitroarginine methyl ester (L-NAME) and high concentration potassium solution were topically applied to simulate the environment after SAH. We irrigated the parietal cortex with artificial cerebrospinal fluid (ACSF), containing L-NAME (1 mM), K+ (35 mM), and Mg2+ (5 mM). Forty-five rats were divided into 3 groups: sham surgery (sham group), L-NAME + [K+]ACSF (control group), and L-NAME + [K+]ACSF + [Mg2+] (Mg group). CSD was induced by topical application with 1 M KCl solution in 3 groups. The effects of Mg administration on CSD and cerebral blood flow were evaluated. Histological brain tissue damage, body weight, and neurological score were assessed at 2 days after insult. RESULTS: Mg solution significantly shortened the total depolarization time, and reduced CSI, histological brain damage, and brain edema compared with those of the control group (P < 0.05). Body weight loss was significantly suppressed in the Mg group (P < 0.05), but neurological score did not improve. CONCLUSIONS: Local application of Mg suppressed CSI and reduced brain damage in a rat SAH-mimicking model. Mg irrigation therapy may be beneficial to suppress brain damage due to CSI after SAH.


Subject(s)
Brain Ischemia/drug therapy , Cortical Spreading Depression/drug effects , Disease Models, Animal , Magnesium Sulfate/administration & dosage , Subarachnoid Hemorrhage/drug therapy , Analgesics/administration & dosage , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Cortical Spreading Depression/physiology , Male , Pharmaceutical Solutions/administration & dosage , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/physiopathology
8.
PLoS One ; 16(8): e0255996, 2021.
Article in English | MEDLINE | ID: mdl-34370788

ABSTRACT

Recent advances in microalgae biotechnology have proven that these microorganisms contain a number of bioactive molecules, that can be used as food additives that help prevent disease. The green microalga Chlorella vulgaris presents several biomolecules, such as lutein and astaxanthin, with antioxidant capacity, which can play a protective role in tissues. In this study, we produced and analyzed a C. vulgaris functional alcoholic beverage (produced using a traditional Brazilian alcoholic beverage, cachaça, and C. vulgaris biomass). Assays were conducted in vitro by radical scavenging tests, and in vivo, by modeling cortical spreading depression in rat brains. Scavenging radical assays showed that consumption of the C. vulgaris alcoholic beverage had a DPPH inhibition of 77.2%. This functional alcoholic beverage at a concentration of 12.5 g L-1 significantly improved cortical spreading depression velocity in the rat brains (2.89 mm min-1), when compared with cachaça alone (3.68 mm min-1) and control (distilled water; 3.25 mm min-1). Moreover, animals that consumed the functional beverage gained less weight than those that consumed just alcohol and the control groups. These findings suggest that the C. vulgaris functional alcoholic beverage plays a protective physiologic role in protecting brain cells from the effects of drinking ethanol.


Subject(s)
Alcoholic Beverages/analysis , Antioxidants/pharmacology , Body Weight/drug effects , Cerebral Cortex/drug effects , Chlorella vulgaris/physiology , Cortical Spreading Depression/drug effects , Animals , Brazil , Male , Rats , Rats, Wistar
9.
Neuropharmacology ; 192: 108612, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34023338

ABSTRACT

Dimethyltryptamine (DMT), an endogenous ligand of sigma-1 receptors (Sig-1Rs), acts against systemic hypoxia, but whether DMT may prevent cerebral ischemic injury is unexplored. Here global forebrain ischemia was created in anesthetized rats and aggravated with the induction of spreading depolarizations (SDs) and subsequent short hypoxia before reperfusion. Drugs (DMT, the selective Sig-1R agonist PRE-084, the Sig-1R antagonist NE-100, or the serotonin receptor antagonist asenapine) were administered intravenously alone or in combination while physiological variables and local field potential from the cerebral cortex was recorded. Neuroprotection and the cellular localization of Sig-1R were evaluated with immunocytochemistry. Plasma and brain DMT content was measured by 2D-LC-HRMS/MS. The affinity of drugs for cerebral Sig-1R was evaluated with a radioligand binding assay. Both DMT and PRE-084 mitigated SDs, counteracted with NE-100. Further, DMT attenuated SD when co-administered with asenapine, compared to asenapine alone. DMT reduced the number of apoptotic and ferroptotic cells and supported astrocyte survival. The binding affinity of DMT to Sig-1R matched previously reported values. Sig-1Rs were associated with the perinuclear cytoplasm of neurons, astrocytes and microglia, and with glial processes. According to these data, DMT may be considered as adjuvant pharmacological therapy in the management of acute cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Cortical Spreading Depression/drug effects , N,N-Dimethyltryptamine/pharmacology , Neurodegenerative Diseases/metabolism , Receptors, sigma/metabolism , Animals , Brain/drug effects , Brain Ischemia/drug therapy , Cortical Spreading Depression/physiology , Dose-Response Relationship, Drug , Male , N,N-Dimethyltryptamine/therapeutic use , Neurodegenerative Diseases/prevention & control , Rats , Rats, Sprague-Dawley , Receptors, sigma/agonists , Sigma-1 Receptor
10.
Sci Rep ; 11(1): 8150, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854148

ABSTRACT

Cortical spreading depolarization (CSD) is a propagating wave of tissue depolarization characterized by a large increase of extracellular potassium concentration and prolonged subsequent electrical silencing of neurons. Waves of CSD arise spontaneously in various acute neurological settings, including migraine aura and ischemic stroke. Recently, we have reported that pan-inhibition of adrenergic receptors (AdRs) facilitates the normalization of extracellular potassium after acute photothrombotic stroke in mice. Here, we have extended that mechanistic study to ask whether AdR antagonists also modify the dynamics of KCl-induced CSD and post-CSD recovery in vivo. Spontaneous neural activity and KCl-induced CSD were visualized by cortex-wide transcranial Ca2+ imaging in G-CaMP7 transgenic mice. AdR antagonism decreased the recurrence of CSD waves and accelerated the post-CSD recovery of neural activity. Two-photon imaging revealed that astrocytes exhibited aberrant Ca2+ signaling after passage of the CSD wave. This astrocytic Ca2+ activity was diminished by the AdR antagonists. Furthermore, AdR pan-antagonism facilitated the normalization of the extracellular potassium level after CSD, which paralleled the recovery of neural activity. These observations add support to the proposal that neuroprotective effects of AdR pan-antagonism arise from accelerated normalization of extracellular K+ levels in the setting of acute brain injury.


Subject(s)
Adrenergic Antagonists/administration & dosage , Cortical Spreading Depression/drug effects , Potassium Chloride/adverse effects , Thrombotic Stroke/drug therapy , Adrenergic Antagonists/pharmacology , Animals , Calcium/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Potassium Chloride/pharmacology , Recovery of Function , Thrombotic Stroke/etiology , Thrombotic Stroke/metabolism , Thrombotic Stroke/physiopathology
11.
Elife ; 102021 04 15.
Article in English | MEDLINE | ID: mdl-33856345

ABSTRACT

Migraine is the sixth most prevalent disease worldwide but the mechanisms that underlie migraine chronicity are poorly understood. Cytoskeletal flexibility is fundamental to neuronal-plasticity and is dependent on dynamic microtubules. Histone-deacetylase-6 (HDAC6) decreases microtubule dynamics by deacetylating its primary substrate, α-tubulin. We use validated mouse models of migraine to show that HDAC6-inhibition is a promising migraine treatment and reveal an undiscovered cytoarchitectural basis for migraine chronicity. The human migraine trigger, nitroglycerin, produced chronic migraine-associated pain and decreased neurite growth in headache-processing regions, which were reversed by HDAC6 inhibition. Cortical spreading depression (CSD), a physiological correlate of migraine aura, also decreased cortical neurite growth, while HDAC6-inhibitor restored neuronal complexity and decreased CSD. Importantly, a calcitonin gene-related peptide receptor antagonist also restored blunted neuronal complexity induced by nitroglycerin. Our results demonstrate that disruptions in neuronal cytoarchitecture are a feature of chronic migraine, and effective migraine therapies might include agents that restore microtubule/neuronal plasticity.


Migraines are a common brain disorder that affects 14% of the world's population. For many people the main symptom of a migraine is a painful headache, often on one side of the head. Other symptoms include increased sensitivity to light or sound, disturbed vision, and feeling sick. These sensory disturbances are called aura and they often occur before the headache begins. One particularly debilitating subset of migraines are chronic migraines, in which patients experience more than 15 headache days per month. Migraine therapies are often only partially effective or poorly tolerated, making it important to develop new drugs for this condition, but unfortunately, little is known about the molecular causes of migraines. To bridge this gap, Bertels et al. used two different approaches to cause migraine-like symptoms in mice. One approach consisted on giving mice nitroglycerin, which dilates blood vessels, produces hypersensitivity to touch, and causes photophobia in both humans and mice. In the second approach, mice underwent surgery and potassium chloride was applied onto the dura, a thick membrane that surrounds the brain. This produces cortical spreading depression, an event that is linked to migraine auras and involves a wave of electric changes in brain cells that slowly propagates across the brain, silencing brain electrical activity for several minutes. Using these approaches, Bertels et al. studied whether causing chronic migraine-like symptoms in mice is associated with changes in the structures of neurons, focusing on the effects of migraines on microtubules. Microtubules are cylindrical protein structures formed by the assembly of smaller protein units. In most cells, microtubules assemble and disassemble depending on what the cell needs. Neurons need stable microtubules to establish connections with other neurons. The experiments showed that provoking chronic migraines in mice led to a reduction in the numbers of connections between different neurons. Additionally, Bertels et al. found that inhibiting HDAC6 (a protein that destabilizes microtubules) reverses the structural changes in neurons caused by migraines and decreases migraine symptoms. The same effects are seen when a known migraine treatment strategy, known as CGRP receptor blockade, is applied. These results suggest that chronic migraines may involve decreased neural complexity, and that the restoration of this complexity by HDAC6 inhibitors could be a potential therapeutic strategy for migraine.


Subject(s)
Brain/drug effects , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Microtubules/drug effects , Migraine Disorders/drug therapy , Neuronal Plasticity/drug effects , Neurons/drug effects , Tubulin/metabolism , Acetylation , Animals , Behavior, Animal/drug effects , Brain/enzymology , Brain/physiopathology , Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Cortical Spreading Depression/drug effects , Disease Models, Animal , Female , Histone Deacetylase 6/metabolism , Male , Mice, Inbred C57BL , Microtubules/enzymology , Microtubules/pathology , Migraine Disorders/chemically induced , Migraine Disorders/enzymology , Migraine Disorders/physiopathology , Neuronal Outgrowth/drug effects , Neurons/enzymology , Neurons/pathology , Nitroglycerin , Pain Perception/drug effects , Pain Threshold/drug effects , Protein Processing, Post-Translational , Receptors, Calcitonin Gene-Related Peptide/drug effects , Receptors, Calcitonin Gene-Related Peptide/metabolism
12.
Behav Brain Res ; 409: 113324, 2021 07 09.
Article in English | MEDLINE | ID: mdl-33915239

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.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Cortical Excitability/physiology , Cortical Spreading Depression/physiology , Hyperalgesia/physiopathology , Hyperhomocysteinemia/complications , Photophobia/physiopathology , Somatosensory Cortex/physiopathology , Animals , Anxiety/etiology , Cortical Spreading Depression/drug effects , Disease Models, Animal , Female , Hyperalgesia/etiology , Hyperhomocysteinemia/chemically induced , Male , Methionine/pharmacology , Migraine with Aura/etiology , Photophobia/etiology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Wistar
13.
Brain ; 144(9): 2863-2878, 2021 10 22.
Article in English | MEDLINE | ID: mdl-33768249

ABSTRACT

Spreading depolarization is a slowly propagating wave of massive cellular depolarization associated with acute brain injury and migraine aura. Genetic studies link depolarizing molecular defects in Ca2+ flux, Na+ current in interneurons, and glial Na+-K+ ATPase with spreading depolarization susceptibility, emphasizing the important roles of synaptic activity and extracellular ionic homeostasis in determining spreading depolarization threshold. In contrast, although gene mutations in voltage-gated potassium ion channels that shape intrinsic membrane excitability are frequently associated with epilepsy susceptibility, it is not known whether epileptogenic mutations that regulate membrane repolarization also modify spreading depolarization threshold and propagation. Here we report that the Kcnq2/Kv7.2 potassium channel subunit, frequently mutated in developmental epilepsy, is a spreading depolarization modulatory gene with significant control over the seizure-spreading depolarization transition threshold, bi-hemispheric cortical expression, and diurnal temporal susceptibility. Chronic DC-band cortical EEG recording from behaving conditional Kcnq2 deletion mice (Emx1cre/+::Kcnq2flox/flox) revealed spontaneous cortical seizures and spreading depolarization. In contrast to the related potassium channel deficient model, Kv1.1-KO mice, spontaneous cortical spreading depolarizations in Kcnq2 cKO mice are tightly coupled to the terminal phase of seizures, arise bilaterally, and are observed predominantly during the dark phase. Administration of the non-selective Kv7.2 inhibitor XE991 to Kv1.1-KO mice partly reproduced the Kcnq2 cKO-like spreading depolarization phenotype (tight seizure coupling and bilateral symmetry) in these mice, indicating that Kv7.2 currents can directly and actively modulate spreading depolarization properties. In vitro brain slice studies confirmed that Kcnq2/Kv7.2 depletion or pharmacological inhibition intrinsically lowers the cortical spreading depolarization threshold, whereas pharmacological Kv7.2 activators elevate the threshold to multiple depolarizing and hypometabolic spreading depolarization triggers. Together these results identify Kcnq2/Kv7.2 as a distinctive spreading depolarization regulatory gene, and point to spreading depolarization as a potentially significant pathophysiological component of KCNQ2-linked epileptic encephalopathy syndromes. Our results also implicate KCNQ2/Kv7.2 channel activation as a potential adjunctive therapeutic target to inhibit spreading depolarization incidence.


Subject(s)
Brain/physiology , Cortical Spreading Depression/physiology , KCNQ2 Potassium Channel/metabolism , Nerve Tissue Proteins/metabolism , Anilides/pharmacology , Animals , Brain/drug effects , Bridged Bicyclo Compounds/pharmacology , Carbamates/pharmacology , Cortical Spreading Depression/drug effects , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/deficiency , Organ Culture Techniques , Phenylenediamines/pharmacology
14.
Food Funct ; 12(7): 3096-3103, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33720258

ABSTRACT

INTRODUCTION: Coconut oil has been considered as a therapeutic alternative in several pathologies, but there is limited information regarding its effects on brain functioning. OBJECTIVE: This study analyzed whether early virgin coconut oil (VCO) supplementation interferes with electrical activity of the adult rat brain and its lipid peroxidation. Moreover, it investigated whether the putative effect on brain electrophysiology could be affected by overnutrition occurring during lactation, and/or by environmental enrichment (EE). Electrophysiology was measured through cortical spreading depression (CSD), a phenomenon related to brain excitability. METHODS: Wistar rats were suckled in litters of either nine or three pups, forming nourished (N) or overnourished (ON) groups, respectively. Between the 7th and 30th days of life, half of the animals in each group received VCO (10 mg kg-1 d-1; by gavage). The other half received an equivalent amount of vehicle (V, 0.009% cremophor). On day 36, animals from both groups were subjected to EE for 4 weeks. At 105 ± 15 days of life, each animal was subjected to CSD recordings and lipid peroxidation analyses. RESULTS: Overnutrition during lactation enhanced body and brain weights. VCO decelerated the CSD propagation velocity (control - 3.57 ± 0.23 mm min-1versus VCO - 3.27 ± 0.18 mm min-1; p < 0.001), regardless of whether subjected to overnourishment or EE exposure. Neither VCO nor EE modified the cerebral lipid peroxidation (p > 0.05). CONCLUSION: VCO supplementation impaired the spreading of CSD, indicating reduction of brain excitability. VCO effects occurred regardless of the nutritional state during lactation.


Subject(s)
Coconut Oil/administration & dosage , Cortical Spreading Depression/drug effects , Animals , Dietary Supplements , Disease Models, Animal , Nutritional Status , Phytotherapy , Rats , Rats, Wistar
15.
Headache ; 61(1): 170-178, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33326598

ABSTRACT

OBJECTIVE: The aim of this study was to determine if the non-convulsant delta-opioid receptor (DOR) agonist, KNT-127, could inhibit migraine-associated endpoints. BACKGROUND: The DOR has been identified as a therapeutic target for migraine. However, the development of delta agonists is limited as some ligands have seizurogenic properties, which may be related to their ability to induce receptor internalization. While both pro- and non-convulsant delta agonists can reduce migraine-associated allodynia, only the proconvulsant agonist, SNC80, has been shown to decrease cortical spreading depression (CSD). It is unclear if the ability of delta agonists to modulate cortical activity is related to the same signaling mechanisms that produce proconvulsant effects. METHODS: The effects of the non-convulsant delta agonist, KNT-127, were examined. Repetitive CSD was induced in female C57BL6/J (n = 6/group) mice by continuous application of KCl and the effect of KNT-127/vehicle (Veh) on both local field potentials and optical intrinsic signals was determined. To assess the effect of KNT-127 on established chronic migraine-associated pain, male and female C57BL6/J mice were treated with nitroglycerin (NTG; 10 mg/kg, ip) every other day for 9 days and tested with KNT-127 (5 mg/kg, sc) or Veh on day 10 (n = 6/group). DOR-enhanced green fluorescent protein mice (n = 4/group) were used to confirm the internalization properties of KNT-127 in the trigeminal ganglia, trigeminal nucleus caudalis, and somatosensory cortex. RESULTS: KNT-127 inhibited CSD events (t(10)  = 3.570, p = 0.0051). In addition, this delta agonist also reversed established cephalic allodynia in the NTG model of chronic migraine (F(1, 20)  = 12.80, p < 0.01). Furthermore, KNT-127 caused limited internalization of DOR in key migraine processing regions. CONCLUSIONS: This study shows that the antimigraine effects of DOR agonists can be separated from their proconvulsant effects. This data provides valuable information for the continued development of delta agonists for the treatment of migraine.


Subject(s)
Analgesics, Opioid/pharmacology , Cortical Spreading Depression/drug effects , Hyperalgesia/drug therapy , Migraine Disorders/drug therapy , Morphinans/pharmacology , Receptors, Opioid, delta/agonists , Analgesics, Opioid/administration & dosage , Animals , Disease Models, Animal , Female , Hyperalgesia/chemically induced , Male , Mice , Mice, Inbred C57BL , Morphinans/administration & dosage , Nitroglycerin/pharmacology , Vasodilator Agents/pharmacology
16.
Nutr Neurosci ; 24(2): 130-139, 2021 Feb.
Article in English | MEDLINE | ID: mdl-31030633

ABSTRACT

Sepsis is a clinical syndrome with high morbidity and mortality. It is characterized by acute inflammatory response and oxidative stress, which is implicated in cerebral dysfunction. Murici (Byrsonimacrassifolia (L.) Kunth) is a fruit rich in antioxidant compounds, which could be an alternative to prevent damage to tissues induced by sepsis . Here, we evaluated the effects of sepsis on the propagation of cortical spreading depression (CSD) and oxidative stress, and tested the action of murici antioxidant extract in prevention against the effect of sepsis. Male Wistar rats (90-210 days, n = 40) were previously supplemented, orogastrically, with murici extract (150 mg/kg/day or 300 mg/kg/day), or an equivalent volume of the vehicle solution, for fifteen days. Then the animals were subjected to experimental sepsis through cecal ligation and perforation (CLP). Subsequently, CSD recordings were obtained and brain oxidative stress was evaluated. Sepsis decelerated CSD and increased the malondialdehyde (MDA) levels in the brain cortex of the animals. In contrast, septic rats that had been previously supplemented with murici antioxidant extract in doses of 150 and 300 mg/kg/day showed an increase in CSD propagation velocity, low levels of MDA and GSH/GSSG ratio and an increase of superoxide dismutase (SOD) activity, regardless of the dose tested. Our results demonstrate that sepsis affects brain excitability and that this effect can be prevented by murici antioxidant extract. The effects of sepsis and/or murici extract on CSD may be due to the oxidative state of the brain.


Subject(s)
Antioxidants/administration & dosage , Cortical Spreading Depression/drug effects , Sepsis/physiopathology , Animals , Fruit/chemistry , Male , Oxidative Stress/drug effects , Plant Extracts/administration & dosage , Rats, Wistar
17.
Nutr Neurosci ; 24(5): 363-370, 2021 May.
Article in English | MEDLINE | ID: mdl-31221041

ABSTRACT

Objectives: Pyridoxine plays a key role in the development of the human nervous system. Several reports suggest that administration of high doses of pyridoxine can be helpful in improving disturbances such as anxiety and pyridoxine-dependent epilepsy, although it has also been associated with a proconvulsive action. In this study, we investigated in developing rats the effects of repeated administration of various doses of pyridoxine on anxiety-like behavior and the brain excitability-related phenomenon known as cortical spreading depression (CSD).Methods: From postnatal day (P) 7 to P27, Wistar rat pups received per gavage pyridoxine hydrochloride (1 mg/kg/day, or 5 mg/kg/day, or 10 mg/kg/day). On P60-70, the animals were tested in the elevated plus maze (EPM) to evaluate anxiety-like behavior. On P71-80, we recorded the CSD (4-hour recording session).Results: Compared with naïve (gavage-free) and saline-treated controls, pyridoxine-treated groups displayed a significant (p < 0.001) increase in CSD propagation velocity and amplitude of the CSD negative direct-current (DC)-shift, and a decrease in the CSD DC-shift duration. These effects were long-lasting and dose-dependent. In the EPM, no significant pyridoxine-associated effect was observed.Discussion: Our data demonstrate a novel action of pyridoxine on an electrical activity-related phenomenon (CSD) in the developing brain, confirming the hypothesis that the chronic treatment with pyridoxine early in life modulates CSD. Data on CSD propagation suggest that pyridoxine at a high dose might act as a prooxidant agent in the developing brain, a hypothesis that deserves further testing.


Subject(s)
Anxiety/physiopathology , Brain/drug effects , Brain/physiopathology , Cortical Spreading Depression/drug effects , Pyridoxine/administration & dosage , Animals , Animals, Newborn , Behavior, Animal/drug effects , Male , Rats, Wistar
18.
Clin Neurol Neurosurg ; 200: 106318, 2021 01.
Article in English | MEDLINE | ID: mdl-33268191

ABSTRACT

BACKGROUND: It is widely known that some patients surgically treated for subdural hematoma (SDH) experience neurologic deficits not clearly explained by the acute brain injury or known sequelae like seizures. There is increasing evidence that cortical spreading depolarization (CSD) may be the cause. A recent article demonstrated that CSD occurred at a rate of 15 % and was associated with neurological deterioration in a subset of patients following chronic subdural hematoma evacuation. Furthermore, CSD can lead to ischemia leading to worsening neurologic deficits. CSD is usually detected on electrocorticography (ECoG) and needs cortical strip electrode placement with equipment and expertise that may not be readily available. CASE DESCRIPTION: We report three cases of patients with subdural hematoma (SDH) not undergoing ECoG in whom CSD was suspected to be the cause of their neurologic deficits post evacuation. Extensive workup including neuroimaging and electroencephalography (EEG) were inconclusive. Patients were subsequently treated with ketamine infusion and had resultant neurological recovery. CONCLUSIONS: Ketamine infusion can help reverse neurologic deficits in patients with SDH in whom the deficits are not explained by neuroimaging or electrographic seizure. CSD is a known phenomenon that can result in neurological injury and must remain in the differential diagnosis of such patients. Though only limited cases are discussed (n = 3), this small case series provides the basis for conducting clinical trials evaluating the efficacy of ketamine in improving functional outcome in brain-injured patients demonstrating evidence of CSD.


Subject(s)
Cortical Spreading Depression/drug effects , Empirical Research , Excitatory Amino Acid Antagonists/administration & dosage , Hematoma, Subdural/drug therapy , Hematoma, Subdural/surgery , Ketamine/administration & dosage , Aged , Aged, 80 and over , Cortical Spreading Depression/physiology , Electroencephalography/drug effects , Female , Hematoma, Subdural/physiopathology , Humans , Male , Middle Aged
19.
Neuropeptides ; 84: 102097, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33059243

ABSTRACT

Despite robust evidence on the role of calcitonin gene-related peptide (CGRP) in migraine via both central and peripheral actions, relatively less is known about how CGRP in the limbic system is involved in migraine progression. This study investigated whether CGRP production machinery exists in the two key limbic regions including hippocampus and amygdala using cortical spreading depression (CSD) as a model of migraine and whether such alteration by CSD is sensitive to N-methyl-d-aspartate (NMDA) receptor regulation in rats. A single or repetitive CSD was induced by topical application of KCl and monitored using electrophysiological methods. The NR2A-containing NMDA receptor antagonist, NVP-AAM077, or its vehicle, was perfused into the contralateral cerebroventricular ventricle of rat. Quantitative PCR was used to measure CGRP mRNA levels in the ipsilateral and contralateral hippocampus and amygdala after CSD events and compared to respective sham treatments. The results showed that neither a single CSD nor repetitive CSD affected CGRP mRNA levels in both the contralateral and ipsilateral hippocampus at 24 h post CSD induction. Differently, significant elevation of CGRP gene expression was observed in the ipsilateral amygdala at 24 h post multiple CSD, but not contralateral side, and not post-single CSD. Further results showed that the CSD-induced CGRP gene expression in the amygdala was markedly reduced by NVP-AAM077 and this reduction corresponded to a reduced cortical susceptibility to CSD in rats. We conclude that repetitive CSD events induce CGRP gene expression in amygdala, which is sensitive to NR2A regulation.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Cortical Spreading Depression/physiology , Gene Expression/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Amygdala/metabolism , Animals , Calcitonin/genetics , Calcitonin/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cortical Spreading Depression/drug effects , Male , Migraine Disorders/metabolism , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects
20.
Prog Brain Res ; 255: 29-67, 2020.
Article in English | MEDLINE | ID: mdl-33008510

ABSTRACT

Migraine is among the most common and disabling neurological diseases in the world. Cortical spreading depression (CSD) is a wave of near-complete depolarization of neurons and glial cells that slowly propagates along the cortex creating the perception of aura. Evidence suggests that CSD can trigger migraine headache. Experimental models of CSD have been considered highly translational as they recapitulate migraine-related phenomena and have been validated for screening migraine therapeutics. Here we outline the essential components of validated experimental models of CSD and provide a comprehensive review of potential modulators and targets against CSD. We further focus on novel interventions that have been recently shown to suppress CSD susceptibility that may lead to therapeutic targets in migraine.


Subject(s)
Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Cortical Spreading Depression/drug effects , Cortical Spreading Depression/physiology , Migraine Disorders/prevention & control , Migraine Disorders/physiopathology , Models, Neurological , Animals , Humans , Migraine with Aura/physiopathology , Migraine with Aura/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...