1
|
Santiago-Castañeda C, Huerta de la Cruz S, Martínez-Aguirre C, Orozco-Suárez SA, Rocha L. Cannabidiol Reduces Short- and Long-Term High Glutamate Release after Severe Traumatic Brain Injury and Improves Functional Recovery. Pharmaceutics 2022; 14:pharmaceutics14081609. [PMID: 36015236 PMCID: PMC9414526 DOI: 10.3390/pharmaceutics14081609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/16/2022] [Accepted: 07/29/2022] [Indexed: 12/04/2022] Open
Abstract
This study aimed to determine if orally administered cannabidiol (CBD) lessens the cortical over-release of glutamate induced by a severe traumatic brain injury (TBI) and facilitates functional recovery. The short-term experiment focused on identifying the optimal oral pretreatment of CBD. Male Wistar rats were pretreated with oral administration of CBD (50, 100, or 200 mg/kg) daily for 7 days. Then, extracellular glutamate concentration was estimated by cortical microdialysis before and immediately after a severe TBI. The long-term experiment focused on evaluating the effect of the optimal treatment of CBD (pre- vs. pre- and post-TBI) 30 days after trauma. Sensorimotor function, body weight, and mortality rate were evaluated. In the short term, TBI induced a high release of glutamate (738% ± 173%; p < 0.001 vs. basal). Oral pretreatment with CBD at all doses tested reduced glutamate concentration but with higher potency at when animals received 100 mg/kg (222 ± 33%, p < 0.01 vs. TBI), an effect associated with a lower mortality rate (22%, p < 0.001 vs. TBI). In the long-term experiment, the TBI group showed a high glutamate concentration (149% p < 0.01 vs. SHAM). In contrast, animals receiving the optimal treatment of CBD (pre- and pre/post-TBI) showed glutamate concentrations like the SHAM group (p > 0.05). This effect was associated with high sensorimotor function improvement. CBD pretreatment, but not pre-/post-treatment, induced a higher body weight gain (39% ± 2.7%, p < 0.01 vs. TBI) and lower mortality rate (22%, p < 0.01 vs. TBI). These results support that orally administered CBD reduces short- and long-term TBI-induced excitotoxicity and facilitated functional recovery. Indeed, pretreatment with CBD was sufficient to lessen the adverse sequelae of TBI.
Collapse
Affiliation(s)
- Cindy Santiago-Castañeda
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City 14330, Mexico; (C.S.-C.); (S.H.d.l.C.); (C.M.-A.)
| | - Saúl Huerta de la Cruz
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City 14330, Mexico; (C.S.-C.); (S.H.d.l.C.); (C.M.-A.)
| | - Christopher Martínez-Aguirre
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City 14330, Mexico; (C.S.-C.); (S.H.d.l.C.); (C.M.-A.)
| | - Sandra Adela Orozco-Suárez
- Unit for Medical Research in Neurological Diseases, Specialties Hospital, National Medical Center SXXI (CMN-SXXI), Mexico City 06720, Mexico;
| | - Luisa Rocha
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City 14330, Mexico; (C.S.-C.); (S.H.d.l.C.); (C.M.-A.)
- Correspondence: ; Tel.: +52-55-5483-2800
| |
Collapse
|
2
|
Santana‐Gomez CE, Engel J, Staba R. Drug-resistant epilepsy and the hypothesis of intrinsic severity: What about the high-frequency oscillations? Epilepsia Open 2021; 7 Suppl 1:S59-S67. [PMID: 34861102 PMCID: PMC9340307 DOI: 10.1002/epi4.12565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 11/19/2022] Open
Abstract
Drug‐resistant epilepsy (DRE) affects approximately one‐third of the patients with epilepsy. Based on experimental findings from animal models and brain tissue from patients with DRE, different hypotheses have been proposed to explain the cause(s) of drug resistance. One is the intrinsic severity hypothesis that posits that drug resistance is an inherent property of epilepsy related to disease severity. Seizure frequency is one measure of epilepsy severity, but frequency alone is an incomplete measure of severity and does not fully explain basic research and clinical studies on drug resistance; thus, other measures of epilepsy severity are needed. One such measure could be pathological high‐frequency oscillations (HFOs), which are believed to reflect the neuronal disturbances responsible for the development of epilepsy and the generation of spontaneous seizures. In this manuscript, we will briefly review the intrinsic severity hypothesis, describe basic and clinical research on HFOs in the epileptic brain, and based on this evidence discuss whether HFOs could be a clinical measure of epilepsy severity. Understanding the mechanisms of DRE is critical for producing breakthroughs in the development and testing of novel strategies for treatment.
Collapse
Affiliation(s)
| | - Jerome Engel
- Department of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
- Brain Research InstituteDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
- Department of NeurobiologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
- Department of Psychiatry and Biobehavioral SciencesDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Richard Staba
- Department of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| |
Collapse
|
3
|
Hsu SK, Hung CF, Yang HC, Weng JR, Wang SJ. TCD, a triterpenoid isolated from wild bitter gourd, reduces synaptosomal release of glutamate and protects against kainic acid-induced neuronal death. Food Funct 2021; 11:9858-9867. [PMID: 33089839 DOI: 10.1039/d0fo02039g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
3β,7β,25-Trihydroxycucurbita-5,23(E)-dien-19-al (TCD) is a triterpenoid isolated from wild bitter gourd that is a common tropical vegetable with neuroprotective effects. Because excessive glutamate release is a major cause of neuronal damage in various neurological disorders, the aims of this study were to examine the effect of TCD on glutamate release in vitro and to examine the effect of TCD in vivo. In rat cerebrocortical synaptosomes, TCD reduced 4-aminopyridine (4-AP)-stimulated glutamate release and Ca2+ concentration elevation, but had no effect on plasma membrane potential. TCD-mediated inhibition of 4-AP-induced glutamate release was dependent on the presence of extracellular calcium; persisted in the presence of the glutamate transporter inhibitor dl-TBOA, P/Q-type Ca2+ channel blocker ω-agatoxin IVA, and intracellular Ca2+-releasing inhibitors dantrolene and CGP37157; and was blocked by the vesicular transporter inhibitor bafilomycin A1 and the N-type Ca2+ channel blocker ω-conotoxin GVIA. Molecular docking studies have demonstrated that TCD binds to N-type Ca2+ channels. TCD-mediated inhibition of 4-AP-induced glutamate release was abolished by the Ca2+-dependent protein kinase C (PKC) inhibitor Go6976, but was unaffected by the Ca2+-independent PKC inhibitor rottlerin. Furthermore, TCD considerably reduced the phosphorylation of PKC, PKCα, and myristoylated alanine-rich C kinase substrate, a major presynaptic substrate for PKC. In a rat model of kainic acid (KA)-induced excitotoxicity, TCD pretreatment substantially attenuated KA-induced neuronal death in the CA3 hippocampal region. These results suggest that TCD inhibits synaptosomal glutamate release by suppressing N-type Ca2+ channels and PKC activity and exerts protective effects against KA-induced excitotoxicity in vivo.
Collapse
Affiliation(s)
- Szu Kai Hsu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| | | | | | | | | |
Collapse
|
4
|
Santiago-Castañeda C, Segovia-Oropeza M, Concha L, Orozco-Suárez SA, Rocha L. Propylparaben Reduces the Long-Term Consequences in Hippocampus Induced by Traumatic Brain Injury in Rats: Its Implications as Therapeutic Strategy to Prevent Neurodegenerative Diseases. J Alzheimers Dis 2020; 82:S215-S226. [PMID: 33185606 DOI: 10.3233/jad-200914] [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] [Indexed: 01/03/2023]
Abstract
BACKGROUND Severe traumatic brain injury (TBI), an important risk factor for Alzheimer's disease, induces long-term hippocampal damage and hyperexcitability. On the other hand, studies support that propylparaben (PPB) induces hippocampal neuroprotection in neurodegenerative diseases. OBJECTIVE Experiments were designed to evaluate the effects of subchronic treatment with PPB on TBI-induced changes in the hippocampus of rats. METHODS Severe TBI was induced using the lateral fluid percussion model. Subsequently, rats received subchronic administration with PPB (178 mg/kg, TBI+PPB) or vehicle (TBI+PEG) daily for 5 days. The following changes were examined during the experimental procedure: sensorimotor dysfunction, changes in hippocampal excitability, as well as neuronal damage and volume. RESULTS TBI+PEG group showed sensorimotor dysfunction (p < 0.001), hyperexcitability (64.2%, p < 0.001), and low neuronal preservation ipsi- and contralateral to the trauma. Magnetic resonance imaging (MRI) analysis revealed lower volume (17.2%; p < 0.01) and great damage to the ipsilateral hippocampus. TBI+PPB group showed sensorimotor dysfunction that was partially reversed 30 days after trauma. This group showed hippocampal excitability and neuronal preservation similar to the control group. However, MRI analysis revealed lower hippocampal volume (p < 0.05) when compared with the control group. CONCLUSION The present study confirms that post-TBI subchronic administration with PPB reduces the long-term consequences of trauma in the hippocampus. Implications of PPB as a neuroprotective strategy to prevent the development of Alzheimer's disease as consequence of TBI are discussed.
Collapse
Affiliation(s)
- Cindy Santiago-Castañeda
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| | - Marysol Segovia-Oropeza
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| | - Luis Concha
- Institute of Neurobiology, National Autonomous University of Mexico, Campus Juriquilla, Queretaro, Mexico
| | - Sandra Adela Orozco-Suárez
- Unit for Medical Research in Neurological Diseases, Specialties Hospital, National Medical Center SXXI (CMN-SXXI), Mexico City, Mexico
| | - Luisa Rocha
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
5
|
Enrique A, Martín P, Sbaraglini ML, Talevi A, Milesi V. Parabens inhibit hNa V 1.2 channels. Biomed Pharmacother 2020; 128:110250. [PMID: 32480218 DOI: 10.1016/j.biopha.2020.110250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/27/2020] [Accepted: 05/10/2020] [Indexed: 10/24/2022] Open
Abstract
Propylparaben, a commonly used antimicrobial preservative, has been reported as an anticonvulsant agent targeting neuronal Na+ channels (NaV). However, the specific features of the NaV channel inhibition by this agent have so far not been extensively studied. Moreover, it is still unclear if it shares this pharmacological activity with other parabens. Here, we fully characterized the mechanism of action of the inhibitory effect that propylparaben and benzylparaben induce on human NaV 1.2 channel isoform (hNaV1.2). We established a first approach to know the parabens structural determinants for this channel inhibition. The parabens effects on hNaV1.2 channel mediated currents were recorded using the patch-clamp whole-cell configuration on hNaV1.2 stably transfected HEK293 cells. Propylparaben induced a typical state-dependent inhibition on hNaV1.2 channel carried current, characterized by a left-shift in the steady-state inactivation curve, a prolongation in the time needed for recovery from fast inactivation and a frequency-dependent blocking behavior. The state-dependent inhibition is increased for butylparaben and benzylparaben and diminished for methylparaben, ethylparaben and p-hydroxybenzoic acid (the major metabolite of parabens hydrolysis). Particularly, butylparaben and benzylparaben shift the steady-state inactivation curve 2- and 3-times more than propylparaben, respectively. Parabens are blockers of hNaV1.2 channels, sharing the mechanism of action of most of sodium channel blocking antiseizure drugs. The potency of this inhibition increases with the size of the lipophilic alcoholic residue of the ester group. These results provide a basis for rational drug design directed to generate new potential anticonvulsant agents.
Collapse
Affiliation(s)
- Andrea Enrique
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, La Plata, Argentina.
| | - Pedro Martín
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, La Plata, Argentina.
| | - María Laura Sbaraglini
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.
| | - Alan Talevi
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.
| | - Verónica Milesi
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, La Plata, Argentina.
| |
Collapse
|
6
|
Barker BS, Spampanato J, McCarren HS, Smolik M, Jackson CE, Hornung EN, Yeung DT, Dudek FE, McDonough JH. Screening for Efficacious Anticonvulsants and Neuroprotectants in Delayed Treatment Models of Organophosphate-induced Status Epilepticus. Neuroscience 2020; 425:280-300. [PMID: 31783100 PMCID: PMC6935402 DOI: 10.1016/j.neuroscience.2019.11.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/30/2019] [Accepted: 11/12/2019] [Indexed: 01/04/2023]
Abstract
Organophosphorus (OP) compounds are deadly chemicals that exert their intoxicating effects through the irreversible inhibition of acetylcholinesterase (AChE). In addition to an excess of peripheral ailments, OP intoxication induces status epilepticus (SE) which if left untreated may lead to permanent brain damage or death. Benzodiazepines are typically the primary therapies for OP-induced SE, but these drugs lose efficacy as treatment time is delayed. The CounterACT Neurotherapeutic Screening (CNS) Program was therefore established by the National Institutes of Health (NIH) to discover novel treatments that may be administered adjunctively with the currently approved medical countermeasures for OP-induced SE in a delayed treatment scenario. The CNS program utilizes in vivo EEG recordings and Fluoro-JadeB (FJB) histopathology in two established rat models of OP-induced SE, soman (GD) and diisopropylfluorophosphate (DFP), to evaluate the anticonvulsant and neuroprotectant efficacy of novel adjunct therapies when administered at 20 or 60 min after the induction of OP-induced SE. Here we report the results of multiple compounds that have previously shown anticonvulsant or neuroprotectant efficacy in other models of epilepsy or trauma. Drugs tested were ganaxolone, diazoxide, bumetanide, propylparaben, citicoline, MDL-28170, and chloroquine. EEG analysis revealed that ganaxolone demonstrated the most robust anticonvulsant activity, whereas all other drugs failed to attenuate ictal activity in both models of OP-induced SE. FJB staining demonstrated that none of the tested drugs had widespread neuroprotective abilities. Overall these data suggest that neurosteroids may represent the most promising anticonvulsant option for OP-induced SE out of the seven unique mechanisms tested here. Additionally, these results suggest that drugs that provide significant neuroprotection from OP-induced SE without some degree of anticonvulsant activity are elusive, which further highlights the necessity to continue screening novel adjunct treatments through the CNS program.
Collapse
Affiliation(s)
- Bryan S Barker
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA.
| | - Jay Spampanato
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Hilary S McCarren
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Melissa Smolik
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Cecelia E Jackson
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Eden N Hornung
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - David T Yeung
- Chemical Countermeasures Research Program, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - John H McDonough
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| |
Collapse
|
7
|
Lu CW, Hung CF, Lin TY, Hsieh TY, Wang SJ. Allicin Inhibits Glutamate Release from Rat Cerebral Cortex Nerve Terminals Through Suppressing Ca2+ Influx and Protein Kinase C Activity. J Med Food 2019; 22:696-702. [DOI: 10.1089/jmf.2018.4337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, Taiwan
- Department of Mechanical Engineering, Yuan Ze University, Taoyuan City, Taiwan
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, Taiwan
- Department of Mechanical Engineering, Yuan Ze University, Taoyuan City, Taiwan
| | - Ting Yang Hsieh
- P.H.D. Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Su Jane Wang
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| |
Collapse
|
8
|
Bellera CL, Talevi A. Quantitative structure-activity relationship models for compounds with anticonvulsant activity. Expert Opin Drug Discov 2019; 14:653-665. [PMID: 31072145 DOI: 10.1080/17460441.2019.1613368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Introduction: Third-generation antiepileptic drugs have seemingly failed to improve the global figures of seizure control and can still be regarded as symptomatic treatments. Quantitative structure-activity relationships (QSAR) can be used to guide hit-to-lead and lead optimization projects and applied to the large-scale virtual screening of chemical libraries. Areas covered: In this review, the authors cover reports on QSAR models related to antiepileptic drugs and drug targets in epilepsy, analyzing whether they refer to classic or non-classic QSAR and if they apply QSAR as a descriptive or predictive approach, among other considerations. The article finally focuses on a more detailed discussion of those predictive studies which include some sort of experimental validation, i.e. papers in which the reported models have been used to identify novel active compounds which have been tested in vitro and/or in vivo. Expert opinion: There are significant opportunities to apply the QSAR methodology to assist the discovery of more efficacious antiepileptic drugs. Considering the intrinsic complexity of the disorder, such applications should focus on state-of-the-art approximations (e.g. systemic, multi-target and multi-scale QSAR as well as ensemble and deep learning) and modeling the effects on novel drug targets and modern screening tools.
Collapse
Affiliation(s)
- Carolina L Bellera
- a Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences , University of La Plata (UNLP) , La Plata, Buenos Aires , Argentina.,b CCT La Plata , Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Buenos Aires , Argentina
| | - Alan Talevi
- a Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences , University of La Plata (UNLP) , La Plata, Buenos Aires , Argentina.,b CCT La Plata , Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Buenos Aires , Argentina
| |
Collapse
|
9
|
Vega-García A, Santana-Gómez CE, Rocha L, Magdaleno-Madrigal VM, Morales-Otal A, Buzoianu-Anguiano V, Feria-Romero I, Orozco-Suárez S. Magnolia officinalis reduces the long-term effects of the status epilepticus induced by kainic acid in immature rats. Brain Res Bull 2019; 149:156-167. [PMID: 30978383 DOI: 10.1016/j.brainresbull.2019.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
During critical periods of neurodevelopment, the immature brain is susceptible to neuronal hyperexcitability, alterations such as hyperthermia, hypoxia, brain trauma or a preexisting neuroinflammatory condition can trigger, promote and prolong epileptiform activity and facilitate the development of epilepsy. The goal of the present study was to evaluate the long-term neuroprotective effects Magnolia officinalis extract, on a model of recurrent status epilepticus (SE) in immature rats. Sprague-Dawley rats were treated with kainic acid (KA) (3 mg/kg, dissolved in saline solution) beginning at day 10 P N every 24 h for five days (10 P N-14PN). Two experimental groups (KA) received two treatments for 10 days (14-24 P N): one group was treated with 300 mg/kg Magnolia Officinalis (MO) (KA-MO), and another was treated with 20 mg/kg of celecoxib (Clbx) (KA-Clbx) as a control drug. A SHAM control group at day 90 P N was established. Seizure susceptibility was analyzed through an after-discharge threshold (ADT) evaluation, and electroencephalographic activity was recorded. The results obtained from the ADT evaluation and the analysis of the electroencephalographic activity under basal conditions showed that the MO and Clbx treatments protected against epileptiform activity, and decreases long-term excitability. All rats in the KA-MO and KA-Clbx groups presented a phase I seizure on the Racine scale, corresponding to the shaking of a wet dog. In contrast, the KA group showed phase V convulsive activity on the Racine scale. Similarly, MO and Clbx exerted neuroprotective effects on hippocampal neurons and reduced gliosis in the same areas. Based on these results, early intervention with MO and Clbx treatments to prevent the inflammatory activity derived from SE in early phases of neurodevelopment exerts neuroprotective effects on epileptogenesis in adult stages.
Collapse
Affiliation(s)
- A Vega-García
- Programa de Doctorado del Departamento de Ciencias Biológicas y de la Salud, UAM-I, Universidad Autónoma Metropolitana Campus Iztapalapa, Ciudad de México, Mexico; Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, IMSS, Ciudad de México, Mexico
| | - C E Santana-Gómez
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados, Tlalpan, Ciudad de México, Mexico
| | - L Rocha
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados, Tlalpan, Ciudad de México, Mexico
| | - V M Magdaleno-Madrigal
- División de Investigación en Neurociencias, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñis", Ciudad de México, Mexico
| | - A Morales-Otal
- Área de Neurociencias. Departamento de Neurohistología y Conducta. Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, Mexico
| | - V Buzoianu-Anguiano
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, IMSS, Ciudad de México, Mexico
| | - I Feria-Romero
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, IMSS, Ciudad de México, Mexico
| | - S Orozco-Suárez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, "Dr. Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, IMSS, Ciudad de México, Mexico.
| |
Collapse
|
10
|
Contreras-García IJ, Pichardo-Macías LA, Santana-Gómez CE, Sánchez-Huerta K, Ramírez-Hernández R, Gómez-González B, Rocha L, Mendoza Torreblanca JG. Differential expression of synaptic vesicle protein 2A after status epilepticus and during epilepsy in a lithium-pilocarpine model. Epilepsy Behav 2018; 88:283-294. [PMID: 30336420 DOI: 10.1016/j.yebeh.2018.08.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/04/2018] [Accepted: 08/17/2018] [Indexed: 11/25/2022]
Abstract
Synaptic vesicle protein 2A (SV2A) has become an attractive target of investigation because of its role in the pathophysiology of epilepsy; SV2A is expressed ubiquitously throughout the brain in all nerve terminals independently of their neurotransmitter content and plays an important but poorly defined role in neurotransmission. Previous studies have shown that modifications in the SV2A protein expression could be a direct consequence of disease severity. Furthermore, these SV2A modifications may depend on specific changes in the nerve tissue following the induction of epilepsy and might be present in both excitatory and inhibitory terminals. Thus, we evaluated SV2A protein expression throughout the hippocampi of lithium-pilocarpine rats after status epilepticus (SE) and during early and late epilepsy. In addition, we determined the γ-aminobutyric acid (GABA)ergic or glutamatergic nature associated with SV2A modifications. Wistar rats were treated with lithium-pilocarpine to induce SE and subsequently were shown to present spontaneous recurrent seizures (SRS). Later, we conducted an exhaustive semi-quantitative analysis of SV2A optical density (OD) throughout the hippocampus by immunohistochemistry. Levels of the SV2A protein were substantially increased in layers formed by principal neurons after SE, mainly because of GABAergic activity. No changes were observed in the early stage of epilepsy. In the late stage of epilepsy, there were minor changes in SV2A OD compared with the robust modifications of SE; however, SV2A protein expression generally showed an increment reaching significant differences in two dendritic layers and hilus, without clear modifications of GABAergic or glutamatergic systems. Our results suggest that the SV2A variations may depend on several factors, such as neuronal activity, and might appear in both excitatory and inhibitory systems depending on the epilepsy stage.
Collapse
Affiliation(s)
- Itzel Jatziri Contreras-García
- Instituto Nacional de Pediatría, Subdirección de Medicina experimental, Laboratorio de Neurociencias, México; Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, México
| | - Luz Adriana Pichardo-Macías
- Instituto Nacional de Pediatría, Subdirección de Medicina experimental, Laboratorio de Neurociencias, México; Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Departamento de Fisiología, México
| | - César Emmanuel Santana-Gómez
- Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Sede Sur México, Departamento de Farmacobiología
| | - Karla Sánchez-Huerta
- Instituto Nacional de Pediatría, Subdirección de Medicina experimental, Laboratorio de Neurociencias, México
| | - Rogelio Ramírez-Hernández
- Instituto Nacional de Pediatría, Subdirección de Medicina experimental, Laboratorio de Neurociencias, México
| | | | - Luisa Rocha
- Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Sede Sur México, Departamento de Farmacobiología
| | | |
Collapse
|
11
|
Valle-Dorado MG, Santana-Gómez CE, Orozco-Suárez SA, Rocha L. Sodium cromoglycate reduces short- and long-term consequences of status epilepticus in rats. Epilepsy Behav 2018; 87:200-206. [PMID: 30115604 DOI: 10.1016/j.yebeh.2018.06.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/18/2018] [Accepted: 06/14/2018] [Indexed: 01/03/2023]
Abstract
Several studies indicate that sodium cromoglycate (CG) induces neuroprotective effects in acute neurological conditions. The present study focused on investigating if the use of CG in rats during the post-status epilepticus (post-SE) period reduces the acute and long-term consequences of seizure activity. Our results revealed that animals that received a single dose of CG (50 mg/kg s.c.: subcutaneously) during the post-SE period showed a lower number of neurons in the process of dying in the dentate gyrus, hilus, cornu ammonis 1 (CA1), and CA3 of the dorsal hippocampus than the rats that received the vehicle. However, this effect was not evident in layers V-VI of the sensorimotor cortex or the lateral-posterior thalamic nucleus. A second experiment showed that animals that received CG subchronically (50 mg/kg s.c. every 12 h for 5 days followed by 24 mg/kg/day s.c. for 14 days using osmotic minipumps) after SE presented fewer generalized convulsive seizures and less neuronal damage in the lateral-posterior thalamic nucleus but not in the hippocampus or cortex. Our data indicate that CG can be used as a therapeutic strategy to reduce short- and long-term neuronal damage in the hippocampus and thalamus, respectively. The data also indicate that CG can reduce the expression of generalized convulsive spontaneous seizures when it is given during the latent period of epileptogenesis.
Collapse
Affiliation(s)
| | | | | | - Luisa Rocha
- Department of Pharmacobiology, Center of Research and Advanced Studies, Mexico City, Mexico.
| |
Collapse
|
12
|
Santana-Gómez CE, Valle-Dorado MG, Domínguez-Valentín AE, Hernández-Moreno A, Orozco-Suárez S, Rocha L. Neuroprotective effects of levetiracetam, both alone and combined with propylparaben, in the long-term consequences induced by lithium-pilocarpine status epilepticus. Neurochem Int 2018; 120:224-232. [PMID: 30213635 DOI: 10.1016/j.neuint.2018.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 08/23/2018] [Accepted: 09/09/2018] [Indexed: 12/27/2022]
Abstract
Status epilepticus (SE) is a neurological condition that frequently induces severe neuronal injury in the hippocampus, subsequent epileptogenesis and pharmacoresistant spontaneous recurrent seizures (SRS). The repeated administration of LEV (a broad-spectrum antiepileptic drug) during the post-SE period does not prevent the subsequent development of SRS. However, this treatment reduces SE-induced neurodegeneration in the hippocampus. Conversely, propylparaben (PPB) is a widely used antimicrobial that blocks voltage-dependent Na+ channels, induces neuroprotection and reduces epileptiform activity in vitro. The present study attempted to determine if the neuroprotective effects induced by LEV are augmented when combined with a sub-effective dose of PPB. Long-term SE-induced consequences (hyperexcitability, high glutamate release, neuronal injury and volume loss) were evaluated in the hippocampus of rats. LEV alone, as well as combined with PPB, did not prevent the occurrence of SRS. However, animals treated with LEV plus PPB showed high prevalence of low frequency oscillations (0.1-4 Hz and 8-90 bands, p < 0.001) and low prevalence of high frequency activity (90-250 bands, p < 0.001) during the interictal period. In addition, these animals presented lower extracellular levels of glutamate, decreased rate of neurodegeneration and a similar hippocampal volume compared to the control conditions. This study's results suggest that LEV associated with PPB could represent a new therapeutic strategy to reduce long-term consequences induced by SE that facilitate pharmacoresistant SRS.
Collapse
Affiliation(s)
| | | | | | | | - Sandra Orozco-Suárez
- Unit for Medical Research in Neurological Diseases, Specialties Hospital, National Medical Center SXXI, Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Luisa Rocha
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico.
| |
Collapse
|
13
|
Bellera CL, Di Ianni ME, Talevi A. The application of molecular topology for ulcerative colitis drug discovery. Expert Opin Drug Discov 2017; 13:89-101. [PMID: 29088918 DOI: 10.1080/17460441.2018.1396314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Although the therapeutic arsenal against ulcerative colitis has greatly expanded (including the revolutionary advent of biologics), there remain patients who are refractory to current medications while the safety of the available therapeutics could also be improved. Molecular topology provides a theoretic framework for the discovery of new therapeutic agents in a very efficient manner, and its applications in the field of ulcerative colitis have slowly begun to flourish. Areas covered: After discussing the basics of molecular topology, the authors review QSAR models focusing on validated targets for the treatment of ulcerative colitis, entirely or partially based on topological descriptors. Expert opinion: The application of molecular topology to ulcerative colitis drug discovery is still very limited, and many of the existing reports seem to be strictly theoretic, with no experimental validation or practical applications. Interestingly, mechanism-independent models based on phenotypic responses have recently been reported. Such models are in agreement with the recent interest raised by network pharmacology as a potential solution for complex disorders. These and other similar studies applying molecular topology suggest that some therapeutic categories may present a 'topological pattern' that goes beyond a specific mechanism of action.
Collapse
Affiliation(s)
- Carolina L Bellera
- a Medicinal Chemistry/Laboratory of Bioactive Research and Development, Department of Biological Sciences, Faculty of Exact Sciences , University of La Plata (UNLP) , La Plata , Buenos Aires , Argentina
| | - Mauricio E Di Ianni
- a Medicinal Chemistry/Laboratory of Bioactive Research and Development, Department of Biological Sciences, Faculty of Exact Sciences , University of La Plata (UNLP) , La Plata , Buenos Aires , Argentina
| | - Alan Talevi
- a Medicinal Chemistry/Laboratory of Bioactive Research and Development, Department of Biological Sciences, Faculty of Exact Sciences , University of La Plata (UNLP) , La Plata , Buenos Aires , Argentina
| |
Collapse
|
14
|
Lara-Valderrábano L, Galván EJ, Rocha L. Propylparaben suppresses epileptiform activity in hippocampal CA1 pyramidal cells in vitro. Epilepsy Res 2017; 136:126-129. [PMID: 28843182 DOI: 10.1016/j.eplepsyres.2017.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 07/18/2017] [Accepted: 08/13/2017] [Indexed: 11/18/2022]
Abstract
Epilepsy is a highly prevalent neurological disorder. Additionally, a percentage of patients do not respond to conventional antiepileptic drugs. Therefore, drugs for epilepsy control are still being developed. In the present study, the effect of propylparaben (PPB) in the epileptiform activity induced by 4-aminopyridine in hippocampal CA1 pyramidal neurons was evaluated using individual recordings in current-clamp mode. Results indicated that PPB suppressed the epileptiform activity in registered neurons. This effect disappeared when PPB was removed from the solution of incubation. In contrast, phenytoin only reduced the firing frequency without abolishing epileptiform activity. Our results indicate that PPB exerts an antiepileptic effect on CA1 pyramidal neurons in vitro. Therefore, PPB may represent an effective antiepileptic compound.
Collapse
Affiliation(s)
| | - Emilio J Galván
- Pharmacobiology Department, Center of Research and Advanced Studies, México City, Mexico
| | - Luisa Rocha
- Pharmacobiology Department, Center of Research and Advanced Studies, México City, Mexico.
| |
Collapse
|