Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.691
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nature ; 631(8021): 663-669, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38961290

RESUMEN

The Warburg effect is a hallmark of cancer that refers to the preference of cancer cells to metabolize glucose anaerobically rather than aerobically1,2. This results in substantial accumulation of lacate, the end product of anaerobic glycolysis, in cancer cells3. However, how cancer metabolism affects chemotherapy response and DNA repair in general remains incompletely understood. Here we report that lactate-driven lactylation of NBS1 promotes homologous recombination (HR)-mediated DNA repair. Lactylation of NBS1 at lysine 388 (K388) is essential for MRE11-RAD50-NBS1 (MRN) complex formation and the accumulation of HR repair proteins at the sites of DNA double-strand breaks. Furthermore, we identify TIP60 as the NBS1 lysine lactyltransferase and the 'writer' of NBS1 K388 lactylation, and HDAC3 as the NBS1 de-lactylase. High levels of NBS1 K388 lactylation predict poor patient outcome of neoadjuvant chemotherapy, and lactate reduction using either genetic depletion of lactate dehydrogenase A (LDHA) or stiripentol, a lactate dehydrogenase A inhibitor used clinically for anti-epileptic treatment, inhibited NBS1 K388 lactylation, decreased DNA repair efficacy and overcame resistance to chemotherapy. In summary, our work identifies NBS1 lactylation as a critical mechanism for genome stability that contributes to chemotherapy resistance and identifies inhibition of lactate production as a promising therapeutic cancer strategy.


Asunto(s)
Proteínas de Ciclo Celular , Resistencia a Antineoplásicos , Ácido Láctico , Proteínas Nucleares , Reparación del ADN por Recombinación , Animales , Femenino , Humanos , Masculino , Ratones , Ácido Anhídrido Hidrolasas/metabolismo , Anaerobiosis , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Inestabilidad Genómica , Ácido Láctico/metabolismo , Lisina/química , Lisina/metabolismo , Lisina Acetiltransferasa 5/metabolismo , Lisina Acetiltransferasa 5/genética , Proteína Homóloga de MRE11/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Organoides , Glucólisis , Terapia Neoadyuvante , L-Lactato Deshidrogenasa/antagonistas & inhibidores , L-Lactato Deshidrogenasa/deficiencia , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Anticonvulsivantes/farmacología
2.
Nat Rev Neurosci ; 25(5): 334-350, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38531962

RESUMEN

Epilepsy remains a major health concern as anti-seizure medications frequently fail, and there is currently no treatment to stop or prevent epileptogenesis, the process underlying the onset and progression of epilepsy. The identification of the pathological processes underlying epileptogenesis is instrumental to the development of drugs that may prevent the generation of seizures or control pharmaco-resistant seizures, which affect about 30% of patients. mTOR signalling and neuroinflammation have been recognized as critical pathways that are activated in brain cells in epilepsy. They represent a potential node of biological convergence in structural epilepsies with either a genetic or an acquired aetiology. Interventional studies in animal models and clinical studies give strong support to the involvement of each pathway in epilepsy. In this Review, we focus on available knowledge about the pathophysiological features of mTOR signalling and the neuroinflammatory brain response, and their interactions, in epilepsy. We discuss mitigation strategies for each pathway that display therapeutic effects in experimental and clinical epilepsy. A deeper understanding of these interconnected molecular cascades could enhance our strategies for managing epilepsy. This could pave the way for new treatments to fill the gaps in the development of preventative or disease-modifying drugs, thus overcoming the limitations of current symptomatic medications.


Asunto(s)
Progresión de la Enfermedad , Epilepsia , Enfermedades Neuroinflamatorias , Serina-Treonina Quinasas TOR , Humanos , Epilepsia/tratamiento farmacológico , Animales , Serina-Treonina Quinasas TOR/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Transducción de Señal/fisiología , Encéfalo/metabolismo , Encéfalo/patología , Anticonvulsivantes/uso terapéutico , Anticonvulsivantes/farmacología
3.
Nature ; 606(7915): 820-826, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35676483

RESUMEN

γ-Aminobutyric acid (GABA) transporter 1 (GAT1)1 regulates neuronal excitation of the central nervous system by clearing the synaptic cleft of the inhibitory neurotransmitter GABA upon its release from synaptic vesicles. Elevating the levels of GABA in the synaptic cleft, by inhibiting GABA reuptake transporters, is an established strategy to treat neurological disorders, such as epilepsy2. Here we determined the cryo-electron microscopy structure of full-length, wild-type human GAT1 in complex with its clinically used inhibitor tiagabine3, with an ordered part of only 60 kDa. Our structure reveals that tiagabine locks GAT1 in the inward-open conformation, by blocking the intracellular gate of the GABA release pathway, and thus suppresses neurotransmitter uptake. Our results provide insights into the mixed-type inhibition of GAT1 by tiagabine, which is an important anticonvulsant medication. Its pharmacodynamic profile, confirmed by our experimental data, suggests initial binding of tiagabine to the substrate-binding site in the outward-open conformation, whereas our structure presents the drug stalling the transporter in the inward-open conformation, consistent with a two-step mechanism of inhibition4. The presented structure of GAT1 gives crucial insights into the biology and pharmacology of this important neurotransmitter transporter and provides blueprints for the rational design of neuromodulators, as well as moving the boundaries of what is considered possible in single-particle cryo-electron microscopy of challenging membrane proteins.


Asunto(s)
Proteínas Transportadoras de GABA en la Membrana Plasmática , Inhibidores de Recaptación de GABA , Ácido gamma-Aminobutírico , Anticonvulsivantes/química , Anticonvulsivantes/farmacología , Microscopía por Crioelectrón , Proteínas Transportadoras de GABA en la Membrana Plasmática/química , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Proteínas Transportadoras de GABA en la Membrana Plasmática/ultraestructura , Inhibidores de Recaptación de GABA/química , Inhibidores de Recaptación de GABA/farmacología , Humanos , Neurotransmisores/metabolismo , Conformación Proteica/efectos de los fármacos , Tiagabina/química , Tiagabina/metabolismo , Tiagabina/farmacología , Ácido gamma-Aminobutírico/metabolismo
4.
Annu Rev Pharmacol Toxicol ; 64: 577-598, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-37788493

RESUMEN

Seizures and other forms of neurovolatility are emerging as druggable prodromal mechanisms that link traumatic brain injury (TBI) to the progression of later dementias. TBI neurotrauma has both acute and long-term impacts on health, and TBI is a leading risk factor for dementias, including chronic traumatic encephalopathy and Alzheimer's disease. Treatment of TBI already considers acute management of posttraumatic seizures and epilepsy, and impressive efforts have optimized regimens of antiepileptic drugs (AEDs) toward that goal. Here we consider that expanding these management strategies could determine which AED regimens best prevent dementia progression in TBI patients. Challenges with this prophylactic strategy include the potential consequences of prolonged AED treatment and that a large subset of patients are refractory to available AEDs. Addressing these challenges is warranted because the management of seizure activity following TBI offers a rare opportunity to prevent the onset or progression of devastating dementias.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Demencia , Epilepsia Postraumática , Humanos , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Epilepsia Postraumática/complicaciones , Epilepsia Postraumática/tratamiento farmacológico , Epilepsia Postraumática/prevención & control , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Demencia/tratamiento farmacológico , Demencia/prevención & control
5.
Proc Natl Acad Sci U S A ; 120(41): e2309773120, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37782796

RESUMEN

Voltage-gated sodium (Nav) channels govern membrane excitability, thus setting the foundation for various physiological and neuronal processes. Nav channels serve as the primary targets for several classes of widely used and investigational drugs, including local anesthetics, antiepileptic drugs, antiarrhythmics, and analgesics. In this study, we present cryogenic electron microscopy (cryo-EM) structures of human Nav1.7 bound to two clinical drugs, riluzole (RLZ) and lamotrigine (LTG), at resolutions of 2.9 Å and 2.7 Å, respectively. A 3D EM reconstruction of ligand-free Nav1.7 was also obtained at 2.1 Å resolution. RLZ resides in the central cavity of the pore domain and is coordinated by residues from repeats III and IV. Whereas one LTG molecule also binds to the central cavity, the other is found beneath the intracellular gate, known as site BIG. Therefore, LTG, similar to lacosamide and cannabidiol, blocks Nav channels via a dual-pocket mechanism. These structures, complemented with docking and mutational analyses, also explain the structure-activity relationships of the LTG-related linear 6,6 series that have been developed for improved efficacy and subtype specificity on different Nav channels. Our findings reveal the molecular basis for these drugs' mechanism of action and will aid the development of novel antiepileptic and pain-relieving drugs.


Asunto(s)
Cannabidiol , Canales de Sodio Activados por Voltaje , Humanos , Anticonvulsivantes/farmacología , Lamotrigina/farmacología , Sodio/metabolismo , Canales de Sodio Activados por Voltaje/química
6.
J Neurochem ; 168(6): 1045-1059, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38291613

RESUMEN

Glial cells play relevant roles in neuroinflammation caused by epilepsy. Elevated hemichannel (HC) activity formed by connexins (Cxs) or pannexin1 (Panx1) largely explains brain dysfunctions commonly caused by neuroinflammation. Glia express HCs formed by Cxs 43, 30, or 26, while glia and neurons both express HCs formed by Panx1. Cx43 HCs allow for the influx of Ca2+, which promotes glial reactivity, enabling the release of the gliotransmitters that contribute to neuronal over-stimulation. Valproate (VPA), an antiseizure medication, has pleiotropic actions on neuronal molecular targets, and their action on glial cell HCs remains elusive. We used HeLa cells transfected with Cx43, Cx30, Cx26, or Panx1 to determine the effect of VPA on HC activity in the brain. VPA slightly increased HC activity under basal conditions, but significantly enhanced it in cells pre-exposed to conditions that promoted HC activity. Furthermore, VPA increased ATP release through Cx43 HCs. The increased HC activity caused by VPA was resistant to washout, being consistent with in silico studies, which predicted the binding site for VPA and Cx43, as well as for Panx1 HCs on the intracellular side, suggesting that VPA first enters through HCs, after which their activity increases.


Asunto(s)
Anticonvulsivantes , Conexinas , Ácido Valproico , Ácido Valproico/farmacología , Humanos , Anticonvulsivantes/farmacología , Conexinas/metabolismo , Células HeLa , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Conexina 43/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Adenosina Trifosfato/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Animales , Epilepsia/metabolismo , Epilepsia/tratamiento farmacológico , Epilepsia/inducido químicamente
7.
Hippocampus ; 34(1): 7-13, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37933097

RESUMEN

There are limited therapeutic options for patients with Dravet syndrome (DS). The equilibrative nucleoside transporters 1 (ENT1) mediate both the influx and efflux of adenosine across the cell membrane exerted beneficial effects in the treatment of epilepsy. This study aimed to evaluate the anticonvulsant effect of the ENT1 inhibitor in an animal model of DS (Scn1aE1099X/+ mice). J7 (5 mg/kg) treatment was efficacious in elevating seizure threshold in Scn1aE1099X/+ mice after hyperthermia exposure. Moreover, the J7 treatment significantly reduced the frequency of spontaneous excitatory post-synaptic currents (sEPSCs, ~35% reduction) without affecting the amplitude in dentate gyrus (DG) granule cells. Pretreatment with the adenosine A1 receptor (A1R) antagonist, DPCPX, abolished the J7 effects on sEPSCs. These observations suggest that the J7 shows an anticonvulsant effect in hyperthermia-induced seizures in Scn1aE1099X/+ mice. This effect possibly acts on presynaptic A1R-mediated signaling modulation in granule cells.


Asunto(s)
Epilepsias Mioclónicas , Epilepsia , Humanos , Ratones , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Nucleósidos/uso terapéutico , Epilepsias Mioclónicas/tratamiento farmacológico , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/metabolismo , Neuronas/metabolismo , Modelos Animales de Enfermedad , Canal de Sodio Activado por Voltaje NAV1.1/genética
8.
Biochem Biophys Res Commun ; 733: 150666, 2024 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-39244848

RESUMEN

Flufenamic acid (FFA) is an anti-inflammatory drug that affects multiple targets and is a widely used research tool in ion channel studies. This pharmacological compound has a low level of selectivity for the transient receptor potential (TRP) channel superfamily, blocking calcium-activated nonselective cation current (ICAN) as well as afterdepolarizations (ADP) induced by it. A number of studies have demonstrated that FFA exerts an anti-epileptic effect in vitro, although the precise mechanism of this effect is not yet identified. The present study used whole-cell patch-clamp recordings and demonstrated that FFA (25 µM) can abolish the generation of seizure-like events (SLE) in entorhinal cortex slices perfused with a 4-aminopyridine-containing solution, depending on the time of application. FFA decreased the temporal summation of synaptic potentials at the onset of SLEs. However, as the epileptiform activity evolved and the SLE onset phase became more abrupt, the blocking effect of FFA diminished. FFA effectively abolished TRP channel-mediated slow ADPs, exerted a weak blockade and slowed the kinetics of GABAa receptor-mediated currents, and did not affect NMDA receptor-mediated evoked currents induced by extracellular stimulation. Although FFA did not directly inhibit NMDA receptor-mediated evoked currents, it decreased the summation of NMDA receptor-mediated potentials in a manner comparable to its effect on the initiation phase of SLE. This suggests that ICAN blockade may be responsible for this effect. Furthermore, our results showed that the selective blocker of melastatin TRP channels (TRPM4) 9-phenanthrol effectively abolished epileptiform activity in a manner analogous to FFA. In contrast, ML-204, the blocker of canonical TRP channels (TRPC), had no discernible effect on this phenomenon. In conclusion, the study demonstrate that FFA abolishes epileptiform activity in the entorhinal cortex by blocking TRPM4 channels and, consequently, decreasing the effectiveness of temporal summation of glutamatergic potentials.


Asunto(s)
Corteza Entorrinal , Ácido Flufenámico , Animales , Corteza Entorrinal/efectos de los fármacos , Corteza Entorrinal/metabolismo , Ácido Flufenámico/farmacología , Masculino , Anticonvulsivantes/farmacología , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Ácido Glutámico/metabolismo , Ratones , Técnicas de Placa-Clamp , Receptores de GABA-A/metabolismo , Epilepsia/metabolismo , Epilepsia/fisiopatología , Epilepsia/tratamiento farmacológico
9.
Biochem Biophys Res Commun ; 725: 150271, 2024 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-38901222

RESUMEN

The R-type voltage-gated calcium channel CaV2.3 is predominantly located in the presynapse and is implicated in distinct types of epileptic seizures. It has consequently emerged as a molecular target in seizure treatment. Here, we determined the cryo-EM structure of the CaV2.3-α2δ1-ß1 complex in the topiramate-bound state at a 3.0 Å resolution. We provide a snapshot of the binding site of topiramate, a widely prescribed antiepileptic drug, on a voltage-gated ion channel. The binding site is located at an intracellular juxtamembrane hydrophilic cavity. Further structural analysis revealed that topiramate may allosterically facilitate channel inactivation. These findings provide fundamental insights into the mechanism underlying the inhibitory effect of topiramate on CaV and NaV channels, elucidating a previously unseen modulator binding site and thus pointing toward a route for the development of new drugs.


Asunto(s)
Anticonvulsivantes , Canales de Calcio Tipo R , Microscopía por Crioelectrón , Topiramato , Anticonvulsivantes/química , Anticonvulsivantes/farmacología , Topiramato/química , Topiramato/farmacología , Humanos , Regulación Alostérica/efectos de los fármacos , Canales de Calcio Tipo R/química , Canales de Calcio Tipo R/metabolismo , Sitios de Unión , Modelos Moleculares , Células HEK293 , Conformación Proteica , Fructosa/química , Fructosa/análogos & derivados , Fructosa/metabolismo , Animales , Proteínas de Transporte de Catión
10.
J Pharmacol Exp Ther ; 391(2): 206-213, 2024 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-38777605

RESUMEN

There is a growing interest in the use of medicinal plants to treat a variety of diseases, and one of the most commonly used medicinal plants globally is Cannabis sativa The two most abundant cannabinoids (Δ9-tetrahydrocannabinol and cannabidiol) have been governmentally approved to treat selected medical conditions; however, the plant produces over 100 cannabinoids, including cannabichromene (CBC). Although the cannabinoids share a common precursor molecule, cannabigerol, they are structurally and pharmacologically unique. These differences may engender differing therapeutic potentials. In this review, we will examine what is currently known about CBC with regards to pharmacodynamics, pharmacokinetics, and receptor profile. We will also discuss the therapeutic areas that have been examined for this cannabinoid, notably antinociceptive, antibacterial, and anti-seizure activities. Finally, we will discuss areas where new research is needed and potential novel medicinal applications for CBC. SIGNIFICANCE STATEMENT: Cannabichromene (CBC) has been suggested to have disparate therapeutic benefits such as anti-inflammatory, anticonvulsant, antibacterial, and antinociceptive effects. Most of the focus on the medical benefits of cannabinoids has been focused on Δ9-tetrahydrocannabinol and cannabidiol. The preliminary studies on CBC indicate that this phytocannabinoid may have unique therapeutic potential that warrants further investigation. Following easier access to hemp, CBC products are commercially available over-the-counter and are being widely utilized with little or no evidence of their safety or efficacy.


Asunto(s)
Cannabinoides , Humanos , Cannabinoides/uso terapéutico , Cannabinoides/farmacología , Animales , Analgésicos/uso terapéutico , Analgésicos/farmacología , Anticonvulsivantes/uso terapéutico , Anticonvulsivantes/farmacología , Antibacterianos/uso terapéutico , Antibacterianos/farmacología
11.
J Pharmacol Exp Ther ; 388(2): 347-357, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-37977809

RESUMEN

Benzodiazepine pharmacoresistance develops when treatment of status epilepticus (SE) is delayed. This response may result from gamma-aminobutyric acid A receptors (GABAAR) internalization that follows prolonged SE; this receptor trafficking results in fewer GABAAR in the synapse to restore inhibition. Increase in synaptic N-methyl-D-aspartate receptors (NMDAR) also occurs in rodent models of SE. Lacosamide, a third-generation antiseizure medication (ASM), acts on the slow inactivation of voltage-gated sodium channels. Another ASM, rufinamide, similarly acts on sodium channels by extending the duration of time spent in the inactivation stage. Combination therapy of the benzodiazepine midazolam, NMDAR antagonist ketamine, and ASMs lacosamide (or rufinamide) was investigated for efficacy against soman (GD)-induced SE and neuropathology. Adult male rats implanted with telemetry transmitters for monitoring electroencephalographic (EEG) activity were exposed to a seizure-inducing dose of GD and treated with an admix of atropine sulfate and HI-6 1 minute later and with midazolam monotherapy or combination therapy 40 minutes after EEG seizure onset. Rats were monitored continuously for seizure activity for two weeks, after which brains were processed for assessment of neurodegeneration, neuronal loss, and neuroinflammatory responses. Simultaneous administration of midazolam, ketamine, and lacosamide (or rufinamide) was more protective against GD-induced SE compared with midazolam monotherapy. In general, lacosamide triple therapy had more positive outcomes on measures of epileptogenesis, EEG power integral, and the number of brain regions protected from neuropathology compared with rats treated with rufinamide triple therapy. Overall, both drugs were well tolerated in these combination models. SIGNIFICANCE STATEMENT: We currently report on improved efficacy of antiseizure medications lacosamide and rufinamide, each administered in combination with ketamine (NMDAR antagonist) and midazolam (benzodiazepine), in combatting soman (GD)-induced seizure, epileptogenesis, and brain pathology over that provided by midazolam monotherapy, or dual therapy of midazolam and lacosamide (or rufinamide) in rats. Administration of lacosamide as adjunct to midazolam and ketamine was particularly effective against GD-induced toxicity. However, protection was incomplete, suggesting the need for further study.


Asunto(s)
Ketamina , Soman , Estado Epiléptico , Triazoles , Ratas , Masculino , Animales , Midazolam/uso terapéutico , Midazolam/farmacología , Lacosamida/efectos adversos , Ketamina/farmacología , Ketamina/uso terapéutico , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Benzodiazepinas , Colinérgicos/efectos adversos , Ácido gamma-Aminobutírico
12.
J Pharmacol Exp Ther ; 388(2): 376-385, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-37770198

RESUMEN

Status epilepticus (SE) is a life-threatening development of self-sustaining seizures that becomes resistant to benzodiazepines when treatment is delayed. Benzodiazepine pharmacoresistance is thought in part to result from internalization of synaptic GABAA receptors, which are the main target of the drug. The naturally occurring neurosteroid allopregnanolone is a therapy of interest against SE for its ability to modulate all isoforms of GABAA receptors. Ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, has been partially effective in combination with benzodiazepines in mitigating SE-associated neurotoxicity. In this study, allopregnanolone as an adjunct to midazolam or midazolam-ketamine combination therapy was evaluated for efficacy against cholinergic-induced SE. Adult male rats implanted with electroencephalographic (EEG) telemetry devices were exposed to the organophosphorus chemical (OP) soman (GD) and treated with an admix of atropine sulfate and HI-6 at 1 minute after exposure followed by midazolam, midazolam-allopregnanolone, or midazolam-ketamine-allopregnanolone 40 minutes after seizure onset. Neurodegeneration, neuronal loss, and neuroinflammation were assessed 2 weeks after GD exposure. Seizure activity, EEG power integral, and epileptogenesis were also compared among groups. Overall, midazolam-ketamine-allopregnanolone combination therapy was effective in reducing cholinergic-induced toxic signs and neuropathology, particularly in the thalamus and hippocampus. Higher dosage of allopregnanolone administered in combination with midazolam and ketamine was also effective in reducing EEG power integral and epileptogenesis. The current study reports that there is a promising potential of neurosteroids in combination with benzodiazepine and ketamine treatments in a GD model of SE. SIGNIFICANCE STATEMENT: Allopregnanolone, a naturally occurring neurosteroid, reduced pathologies associated with soman (GD) exposure such as epileptogenesis, neurodegeneration, and neuroinflammation, and suppressed GD-induced toxic signs when used as an adjunct to midazolam and ketamine in a delayed treatment model of soman-induced status epilepticus (SE) in rats. However, protection was incomplete, suggesting that further studies are needed to identify optimal combinations of antiseizure medications and routes of administration for maximal efficacy against cholinergic-induced SE.


Asunto(s)
Ketamina , Neuroesteroides , Soman , Estado Epiléptico , Ratas , Masculino , Animales , Midazolam/farmacología , Midazolam/uso terapéutico , Ketamina/farmacología , Ketamina/uso terapéutico , Pregnanolona/efectos adversos , Soman/toxicidad , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Enfermedades Neuroinflamatorias , Neuroesteroides/uso terapéutico , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Benzodiazepinas , Colinérgicos/efectos adversos , Receptores de GABA-A , Ácido gamma-Aminobutírico
13.
J Pharmacol Exp Ther ; 391(1): 104-118, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39060163

RESUMEN

Human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND) affects nearly half of the 39 million people living with HIV. HAND symptoms range from subclinical cognitive impairment to dementia; the mechanisms that underlie HAND remain unclear and there is no treatment. The HIV protein transactivator of transcription (TAT) is thought to contribute to HAND because it persists in the central nervous system and elicits neurotoxicity in animal models. Network hyperexcitability is associated with accelerated cognitive decline in neurodegenerative disorders. Here we show that the antiepileptic drug levetiracetam (LEV) attenuated aberrant excitatory synaptic transmission, protected synaptic plasticity, reduced seizure susceptibility, and preserved cognition in inducible TAT (iTAT) transgenic male mice. iTAT mice had an increased frequency of spontaneous excitatory postsynaptic currents in hippocampal slice recordings and impaired long-term potentiation, a form of synaptic plasticity that underlies learning and memory. Two-week administration of LEV by osmotic minipump prevented both impairments. Kainic acid administered to iTAT mice induced a higher maximum behavioral seizure score, longer seizure duration, and shorter latency to first seizure, consistent with a lower seizure threshold. LEV treatment prevented these in vivo signs of hyperexcitability. Lastly, in the Barnes maze, iTAT mice required more time to reach the goal, committed more errors, and received lower cognitive scores relative to iTAT mice treated with LEV. Thus, TAT expression drives functional deficits, suggesting a causative role in HAND. As LEV not only prevented aberrant synaptic activity in iTAT mice but also prevented cognitive dysfunction, it may provide a promising pharmacological approach to the treatment of HAND. SIGNIFICANCE STATEMENT: Approximately half of people living with human immunodeficiency virus (HIV) also suffer from HIV-associated neurocognitive disorder (HAND), for which there is no treatment. The HIV protein transactivator of transcription (TAT) causes toxicity that is thought to contribute to HAND. Here, the antiepileptic drug levetiracetam (LEV) prevented synaptic and cognitive impairments in a TAT-expressing mouse. LEV is widely used to treat seizures and is well-tolerated in humans, including those with HIV. This study supports further investigation of LEV-mediated neuroprotection in HAND.


Asunto(s)
Complejo SIDA Demencia , Cognición , Levetiracetam , Ratones Transgénicos , Piracetam , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Animales , Levetiracetam/farmacología , Levetiracetam/uso terapéutico , Ratones , Masculino , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Piracetam/análogos & derivados , Piracetam/farmacología , Piracetam/uso terapéutico , Cognición/efectos de los fármacos , Complejo SIDA Demencia/tratamiento farmacológico , Modelos Animales de Enfermedad , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , VIH-1/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Convulsiones/tratamiento farmacológico , Plasticidad Neuronal/efectos de los fármacos , Ratones Endogámicos C57BL , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
14.
Toxicol Appl Pharmacol ; 484: 116870, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38395364

RESUMEN

The development of refractory status epilepticus (SE) following sarin intoxication presents a therapeutic challenge. Here, we evaluated the efficacy of delayed combined double or triple treatment in reducing abnormal epileptiform seizure activity (ESA) and the ensuing long-term neuronal insult. SE was induced in rats by exposure to 1.2 LD50 sarin followed by treatment with atropine and TMB4 (TA) 1 min later. Double treatment with ketamine and midazolam or triple treatment with ketamine, midazolam and levetiracetam was administered 30 min post-exposure, and the results were compared to those of single treatment with midazolam alone or triple treatment with ketamine, midazolam, and valproate, which was previously shown to ameliorate this neurological insult. Toxicity and electrocorticogram activity were monitored during the first week, and behavioral evaluations were performed 2 weeks post-exposure, followed by biochemical and immunohistopathological analyses. Both double and triple treatment reduced mortality and enhanced weight recovery compared to TA-only treatment. Triple treatment and, to a lesser extent, double treatment significantly ameliorated the ESA duration. Compared to the TA-only or the TA+ midazolam treatment, both double and triple treatment reduced the sarin-induced increase in the neuroinflammatory marker PGE2 and the brain damage marker TSPO and decreased gliosis, astrocytosis and neuronal damage. Finally, both double and triple treatment prevented a change in behavior, as measured in the open field test. No significant difference was observed between the efficacies of the two triple treatments, and both triple combinations completely prevented brain injury (no differences from the naïve rats). Delayed double and, to a greater extent, triple treatment may serve as an efficacious delayed therapy, preventing brain insult propagation following sarin-induced refractory SE.


Asunto(s)
Lesiones Encefálicas , Ketamina , Agentes Nerviosos , Estado Epiléptico , Ratas , Animales , Sarín/toxicidad , Agentes Nerviosos/toxicidad , Midazolam/farmacología , Midazolam/uso terapéutico , Ratas Sprague-Dawley , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Colinérgicos/efectos adversos , Lesiones Encefálicas/inducido químicamente
15.
Epilepsia ; 65(8): 2470-2482, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39119787

RESUMEN

OBJECTIVE: Given its key homeostatic role affecting mitochondria, ionotropic and metabotropic receptors, and voltage-gated ion channels, sigma-1 receptor (Sig1R) represents an interesting target for epilepsy management. Antiseizure effects of the positive allosteric modulator E1R have already been reported in acute seizure models. Although modulation of serotonergic neurotransmission is considered the main mechanism of action of fenfluramine, its interaction with Sig1R may be of additional relevance. METHODS: To further explore the potential of Sig1R as a target, we assessed the efficacy and tolerability of E1R and fenfluramine in two chronic mouse models, including an amygdala kindling paradigm and the intrahippocampal kainate model. The relative contribution of the interaction with Sig1R was analyzed using combination experiments with the Sig1R antagonist NE-100. RESULTS: Whereas E1R exerted pronounced dose-dependent antiseizure effects at well-tolerated doses in fully kindled mice, only limited effects were observed in response to fenfluramine, without a clear dose dependency. In the intrahippocampal kainate model, E1R failed to influence electrographic seizure activity. In contrast, fenfluramine significantly reduced the frequency of electrographic seizure events and their cumulative duration. Pretreatment with NE-100 reduced the effects of E1R and fenfluramine in the kindling model. Surprisingly, pre-exposure to NE-100 in the intrahippocampal kainate model rather enhanced and prolonged fenfluramine's antiseizure effects. SIGNIFICANCE: In conclusion, the kindling data further support Sig1R as an interesting target for novel antiseizure medications. However, it is necessary to further explore the preclinical profile of E1R in chronic epilepsy models with spontaneous seizures. Despite the rather limited effects in the kindling paradigm, the findings from the intrahippocampal kainate model suggest that it is of interest to further assess a possible broad-spectrum potential of fenfluramine.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia , Fenfluramina , Excitación Neurológica , Receptores sigma , Receptor Sigma-1 , Animales , Receptores sigma/antagonistas & inhibidores , Receptores sigma/efectos de los fármacos , Ratones , Excitación Neurológica/efectos de los fármacos , Fenfluramina/farmacología , Epilepsia/tratamiento farmacológico , Masculino , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Relación Dosis-Respuesta a Droga , Piperazinas/farmacología , Piperazinas/uso terapéutico , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiopatología , Hipocampo/efectos de los fármacos , Enfermedad Crónica , Ácido Kaínico/farmacología , Ratones Endogámicos C57BL
16.
Epilepsia ; 65(10): 2923-2934, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39140981

RESUMEN

OBJECTIVE: Pharmacological activation of neuronal Kv7 channels by the antiepileptic drug retigabine (RTG; ezogabine) has been proven effective in treating partial epilepsy. However, RTG was withdrawn from the market due to the toxicity caused by its phenazinium dimer metabolites, leading to peripheral skin discoloration and retinal abnormalities. To address the undesirable metabolic properties of RTG and prevent the formation of phenazinium dimers, we made chemical modifications to RTG, resulting in a new RTG derivative, 1025c, N,N'-{4-[(4-fluorobenzyl) (prop-2-yn-1-yl)amino]-1,2-phenylene}bis(3,3-dimethylbutanamide). METHODS: Whole-cell recordings were used to evaluate Kv7 channel openers. Site-directed mutagenesis and molecular docking were adopted to investigate the molecular mechanism underlying 1025c and Kv7.2 interactions. Mouse seizure models of maximal electroshock (MES), subcutaneous pentylenetetrazol (scPTZ), and PTZ-induced kindling were utilized to test compound antiepileptic activity. RESULTS: The novel compound 1025c selectively activates whole-cell Kv7.2/7.3 currents in a concentration-dependent manner, with half-maximal effective concentration of .91 ± .17 µmol·L-1. The 1025c compound also causes a leftward shift in Kv7.2/7.3 current activation toward a more hyperpolarized membrane potential, with a shift of the half voltage of maximal activation (ΔV1/2) of -18.6 ± 3.0 mV. Intraperitoneal administration of 1025c demonstrates dose-dependent antiseizure activities in assays of MES, scPTZ, and PTZ-induced kindling models. Moreover, through site-directed mutagenesis combined with molecular docking, a key residue Trp236 has been identified as critical for 1025c-mediated activation of Kv7.2 channels. Photostability experiments further reveal that 1025c is more photostable than RTG and is unable to dimerize. SIGNIFICANCE: Our findings demonstrate that 1025c exhibits potent and selective activation of neuronal Kv7 channels without being metabolized to phenazinium dimers, suggesting its developmental potential as an antiseizure agent for therapy.


Asunto(s)
Anticonvulsivantes , Carbamatos , Canal de Potasio KCNQ2 , Fenilendiaminas , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/química , Ratones , Fenilendiaminas/farmacología , Carbamatos/farmacología , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ2/metabolismo , Convulsiones/tratamiento farmacológico , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Masculino , Humanos , Simulación del Acoplamiento Molecular , Pentilenotetrazol/toxicidad , Técnicas de Placa-Clamp , Electrochoque , Células HEK293 , Modelos Animales de Enfermedad , Mutagénesis Sitio-Dirigida , Relación Dosis-Respuesta a Droga , Ratones Endogámicos C57BL , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/metabolismo
17.
Neurochem Res ; 49(9): 2379-2392, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38837094

RESUMEN

Epilepsy is a common neurological disorder, and the exploration of potential therapeutic drugs for its treatment is still ongoing. Vitamin D has emerged as a promising treatment due to its potential neuroprotective effects and anti-epileptic properties. This study aimed to investigate the effects of vitamin D on epilepsy and neuroinflammation in juvenile mice using network pharmacology and molecular docking, with a focus on the mammalian target of rapamycin (mTOR) signaling pathway. Experimental mouse models of epilepsy were established through intraperitoneal injection of pilocarpine, and in vitro injury models of hippocampal neurons were induced by glutamate (Glu) stimulation. The anti-epileptic effects of vitamin D were evaluated both in vivo and in vitro. Network pharmacology and molecular docking analysis were used to identify potential targets and regulatory pathways of vitamin D in epilepsy. The involvement of the mTOR signaling pathway in the regulation of mouse epilepsy by vitamin D was validated using rapamycin (RAPA). The levels of inflammatory cytokines (TNF-α, IL-1ß, and IL-6) were assessed by enzyme-linked immunosorbent assay (ELISA). Gene and protein expressions were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. The terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) staining was used to analyze the apoptosis of hippocampal neurons. In in vivo experiments, vitamin D reduced the Racine scores of epileptic mice, prolonged the latency of epilepsy, and inhibited the production of TNF-α, IL-1ß, and IL-6 in the hippocampus. Furthermore, network pharmacology analysis identified RAF1 as a potential target of vitamin D in epilepsy, which was further confirmed by molecular docking analysis. Additionally, the mTOR signaling pathway was found to be involved in the regulation of mouse epilepsy by vitamin D. In in vitro experiments, Glu stimulation upregulated the expressions of RAF1 and LC3II/LC3I, inhibited mTOR phosphorylation, and induced neuronal apoptosis. Mechanistically, vitamin D activated the mTOR signaling pathway and alleviated mouse epilepsy via RAF1, while the use of the pathway inhibitor RAPA reversed this effect. Vitamin D alleviated epilepsy symptoms and neuroinflammation in juvenile mice by activating the mTOR signaling pathway via RAF1. These findings provided new insights into the molecular mechanisms underlying the anti-epileptic effects of vitamin D and further supported its use as an adjunctive therapy for existing anti-epileptic drugs.


Asunto(s)
Epilepsia , Simulación del Acoplamiento Molecular , Farmacología en Red , Proteínas Proto-Oncogénicas c-raf , Transducción de Señal , Serina-Treonina Quinasas TOR , Vitamina D , Animales , Serina-Treonina Quinasas TOR/metabolismo , Epilepsia/tratamiento farmacológico , Epilepsia/metabolismo , Transducción de Señal/efectos de los fármacos , Ratones , Vitamina D/farmacología , Vitamina D/uso terapéutico , Masculino , Proteínas Proto-Oncogénicas c-raf/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
18.
PLoS Comput Biol ; 19(3): e1010919, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36867652

RESUMEN

The ability of neural circuits to integrate information over time and across different cortical areas is believed an essential ingredient for information processing in the brain. Temporal and spatial correlations in cortex dynamics have independently been shown to capture these integration properties in task-dependent ways. A fundamental question remains if temporal and spatial integration properties are linked and what internal and external factors shape these correlations. Previous research on spatio-temporal correlations has been limited in duration and coverage, thus providing only an incomplete picture of their interdependence and variability. Here, we use long-term invasive EEG data to comprehensively map temporal and spatial correlations according to cortical topography, vigilance state and drug dependence over extended periods of time. We show that temporal and spatial correlations in cortical networks are intimately linked, decline under antiepileptic drug action, and break down during slow-wave sleep. Further, we report temporal correlations in human electrophysiology signals to increase with the functional hierarchy in cortex. Systematic investigation of a neural network model suggests that these dynamical features may arise when dynamics are poised near a critical point. Our results provide mechanistic and functional links between specific measurable changes in the network dynamics relevant for characterizing the brain's changing information processing capabilities.


Asunto(s)
Anticonvulsivantes , Vigilia , Humanos , Anticonvulsivantes/farmacología , Encéfalo/fisiología , Mapeo Encefálico/métodos , Imagen por Resonancia Magnética/métodos
19.
Exp Mol Pathol ; 139: 104921, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39096892

RESUMEN

Seizures are neurological disorders triggered by an imbalance in the activity of excitatory and inhibitory neurotransmitters in the brain. When triggered chronically, this imbalance can lead to epilepsy. Critically, many of the affected individuals are refractory to treatment. Given this, anti-inflammatory drugs, in particular glucocorticoids, have been considered as a potential antiepileptogenic therapy. Glucocorticoids are currently used in the treatment of refractory patients, although there have been contradictory results in terms of their use in association with antiepileptic drugs, which reinforces the need for a more thorough investigation of their effects. In this context, the present study evaluated the effects of dexamethasone (DEX, 0.6 mg/kg) on the electroencephalographic (EEG) and histopathological parameters of male Wistar rats submitted to acute seizure induced by pentylenetetrazol (PTZ). The EEG monitoring revealed that DEX reduced the total brainwave power, in comparison with PTZ, in 12 h after the convulsive episode, exerting this effect in up to 36 h (p < 0.05 for all comparisons). An increase in the accommodation of the oscillations of the delta, alpha, and gamma frequencies was also observed from the first 12 h onwards, with the accommodation of the theta frequency occurring after 36 h, and that of the beta frequency 24 h after the seizure. The histopathological analyses showed that the CA3 region and hilum of the hippocampus suffered cell loss after the PTZ-induced seizure (control vs. PTZ, p < 0.05), although DEX was not able to protect these regions against cell death (PTZ vs. DEX + PTZ, p > 0.05). While DEX did not reverse the cell damage caused by PTZ, the data indicate that DEX has beneficial properties in the EEG analysis, which makes it a promising candidate for the attenuation of the epileptiform wave patterns that can precipitate refractory seizures.


Asunto(s)
Ondas Encefálicas , Dexametasona , Electroencefalografía , Pentilenotetrazol , Ratas Wistar , Convulsiones , Animales , Dexametasona/farmacología , Dexametasona/efectos adversos , Pentilenotetrazol/toxicidad , Convulsiones/tratamiento farmacológico , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Masculino , Ratas , Ondas Encefálicas/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Anticonvulsivantes/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/fisiopatología
20.
Org Biomol Chem ; 22(5): 1027-1033, 2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38193622

RESUMEN

γ-Aminobutyric acid (GABA) and GABA derivatives have attracted increased attention over the years in the fields of medicinal chemistry and chemical biology due to their interesting biological properties and synthetic relevance. Here, we report a short synthetic route to γ-(het)aryl- and γ-alkenyl-γ-aminobutyric acids, including the antiepileptic drug vigabatrin, from readily available donor-acceptor cyclopropanes and ammonia or methylamine. This protocol includes a facile synthesis of 2-oxopyrrolidine-3-carboxamides and their acid hydrolysis to γ-aryl- or γ-alkenyl-substituted GABAs, which can serve as perspective building blocks for the synthesis of various GABA-based N-heterocycles and bioactive compounds.


Asunto(s)
Vigabatrin , Ácido gamma-Aminobutírico , Anticonvulsivantes/farmacología , Vigabatrin/farmacología , Pirrolidinas/química , Pirrolidinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA