Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Neurobiol Dis ; : 106592, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38971479

RESUMEN

Failure to recover from repeated hypercapnia and hypoxemia challenges caused by severe GCS and postictal apneas may contribute to sudden unexpected death in epilepsy (SUDEP). Our previous studies found orexinergic dysfunction contributes to respiratory abnormalities in a preclinical model of SUDEP, Kcna1-/-mice. Here, we developed two gas challenges consisting of repeated HH exposures and used wholebody plethysmography to determine whether Kcna1-/-mice would have detrimental ventilatory responses. Kcna1-/- mice exhibited an elevated ventilatory response to a mild repeated hypercapnia-hypoxia (HH) challenge compared to WT. Moreover, 71% of Kcna1-/- mice failed to survive a severe repeated HH challenge, whereas all WT mice recovered. We next determined whether orexin was involved in these differences. Pretreatment of Kcna1-/- mice with a dual orexin receptor antagonist rescued the ventilatory response during the mild challenge and all subjects survived the severe challenge. In ex vivo extracellular recordings in the lateral hypothalamus of coronal brain slices, we found reducing pH either inhibits or stimulates putative orexin neurons similar to other chemosensitive neurons; however, a significantly greater percentage of putative orexin neurons from Kcna1-/-mice were stimulated and the magnitude of stimulation was increased resulting in augmentation of the calculated chemosensitivity index relative to WT. Collectively, our data suggest that increased chemosensitive activity of orexin neurons may be pathologic in the Kcna1-/- mouse model of SUDEP, and contribute to elevated ventilatory responses. Our data suggest that individuals at high risk for SUDEP may be more sensitive to HH challenges, whether induced by seizures or other means; and the depth and length of the HH exposure could dictate the probability of survival.

2.
J Reconstr Microsurg ; 39(1): 20-26, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35477114

RESUMEN

BACKGROUND: Perforators are typically found in rows in the deep inferior epigastric perforator (DIEP) flap. As methods to assess flap perfusion continue to improve, surgeons may be more likely to select perforators traditionally avoided. The purpose of this article is to describe clinical outcomes based on row and number of perforators to reevaluate flap and abdominal donor site morbidity. METHODS: A retrospective analysis was performed on patients who underwent breast reconstruction with DIEP flaps by four microsurgeons from 2013 to 2020. The row and number of perforators were determined from operative reports. Chi-square and t-test or nonparametric Fisher's exact test and Wilcoxon two-sample test were used for discrete and continuous variable, respectively, as applicable. Logistic regression was used for multivariable analyses. RESULTS: Of 628 flaps, 305 were medial row (58.7%), 159 were lateral row (30.6%), and 55 had both rows (10.6%). Partial flap loss was higher in both rows (p = 0.003). Fat necrosis was higher with medial (p = 0.03) and both rows (p = 0.01) when compared with lateral using multivariable analysis. Hernia or bulge was higher in lateral row flaps (lateral: 8/157, 5.1%; medial, 5/299, 1.7%; both, 0/55; p = 0.05); however, mesh was more commonly used in both row flaps (p = 0.05). There was no difference in fat necrosis or abdominal morbidity between single and multiple perforators. CONCLUSION: There was no difference in fat necrosis based on the number or row of perforators. The lateral row provides adequate perfusion but may be associated with an elevated risk of hernia or bulge. Patients may benefit from mesh, especially when both rows are dissected.


Asunto(s)
Necrosis Grasa , Mamoplastia , Colgajo Perforante , Humanos , Colgajo Perforante/cirugía , Estudios Retrospectivos , Mamoplastia/métodos , Arterias Epigástricas/cirugía , Hernia
3.
Epilepsia ; 62(8): e123-e128, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34231878

RESUMEN

There is growing evidence for the disease-modifying potential of metabolic therapies, including the ketogenic diet (KD), which is used to treat medically intractable epilepsy. However, it remains unclear whether the KD exerts direct effects on histopathological changes in epileptic brain, or whether the changes are a consequence of diet-induced reduction in seizure activity. Here, we used unbiased stereological techniques to quantify the seizure-induced reduction in cell number in the CA1 region of the hippocampus of epileptic Kcna1-null mice and compared the effects of the KD with that of phenobarbital (PB), a widely employed anti-seizure drug. Our data suggest that the anti-seizure activity of the KD or PB was similar. However, CA1 cell numbers of KD-treated hippocampi were not significantly different from those seen in wild-type (WT) mice, whereas CA1 cell counts in standard diet and PB-treated Kcna1-null mice were 23% and 31% lower than WT animals, respectively. These results support the notion that structural protection of cells may involve more than seizure attenuation, and that the KD engages mechanisms that also promote or restore hippocampal morphological integrity.


Asunto(s)
Dieta Cetogénica , Epilepsia , Convulsiones , Animales , Recuento de Células , Epilepsia/dietoterapia , Epilepsia/genética , Canal de Potasio Kv.1.1/genética , Ratones , Ratones Noqueados , Convulsiones/genética
4.
Epilepsia ; 59(1): 92-105, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29193044

RESUMEN

OBJECTIVE: Chronic sleep deficiency is associated with early mortality. In the epileptic population, there is a higher prevalence of sleep disorders, and individuals with severe refractory epilepsy are at greater risk of premature mortality than the general population. Sudden unexpected death in epilepsy affects 1:1000 cases of epilepsy each year. Ketogenic diet (KD) treatment is one of the few effective options for refractory seizures. Despite KD reducing seizures and increasing longevity in Kv1.1 knockout (KO) mice, they still succumb to sudden death. This study aims to determine whether (1) the rest profiles of KO and KD-treated KO (KOKD) mice resemble each other as a function of either age or proximity to death and (2) the timing of death correlates with acute or chronic changes in rest. METHODS: Noninvasive actimetry was used to monitor rest throughout the lives of KO and wild-type (WT) littermates administered standard diet or KD. RESULTS: As KO mice age, rest is reduced (P < .0001). Rest is significantly improved in KDKO mice (P < .0001), resembling WT values at several ages. When age is removed as a variable and data are realigned to the day of death, the rest profiles of KO and KOKD groups worsen to similar degrees as a function of proximity to death. The amount of rest acutely is not sensitive to the timing of death, whereas chronic rest deficiency profiles (10-15 days prior to death) of both groups were indistinguishable. Chronic accumulation of rest deficiency over the final 15 days was associated with 75% of deaths. SIGNIFICANCE: Our data suggest that the accumulated rest deficiency is associated with sudden death in Kv1.1 KO mice. These data (1) support the proposed clinical hypothesis that chronic sleep deficiency may be associated with early mortality in epileptic patients and (2) warrant future preclinical and clinical studies on sleep monitoring in epileptic patients.


Asunto(s)
Muerte Súbita , Epilepsia/genética , Epilepsia/fisiopatología , Canal de Potasio Kv.1.1/deficiencia , Privación de Sueño/genética , Privación de Sueño/fisiopatología , Actigrafía , Factores de Edad , Animales , Dieta Cetogénica/métodos , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia/dietoterapia , Canal de Potasio Kv.1.1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Privación de Sueño/dietoterapia , Telemetría
5.
Epilepsia ; 59(2): 345-357, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29327348

RESUMEN

OBJECTIVE: Increased breathing rate, apnea, and respiratory failure are associated with sudden unexpected death in epilepsy (SUDEP). We recently demonstrated the progressive nature of epilepsy and mortality in Kcna1-/- mice, a model of temporal lobe epilepsy and SUDEP. Here we tested the hypothesis that respiratory dysfunction progresses with age in Kcna1-/- mice, thereby increasing risk of respiratory failure and sudden death (SD). METHODS: Respiratory parameters were determined in conscious mice at baseline and following increasing doses of methacholine (MCh) using noninvasive airway mechanics (NAM) systems. Kcna1+/+ , Kcna1+/- , and Kcna1-/- littermates were assessed during 3 age ranges when up to ~30%, ~55%, and ~90% of Kcna1-/- mice have succumbed to SUDEP: postnatal day (P) 32-36, P40-46, and P48-56, respectively. Saturated arterial O2 (SaO2 ) was determined with pulse oximetry. Lung and brain tissues were isolated and Kcna1 gene and protein expression were evaluated by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) and Western blot techniques. Airway smooth muscle responsiveness was assessed in isolated trachea exposed to MCh. RESULTS: Kcna1-/- mice experienced an increase in basal respiratory drive, chronic oxygen desaturation, frequent apnea-hypopnea (A-H), an atypical breathing sequence of A-H-tachypnea-A-H, increased tidal volume, and hyperventilation induced by MCh. The MCh-provoked hyperventilation was dramatically attenuated with age. Of interest, only Kcna1-/- mice developed seizures following exposure to MCh. Seizures were provoked by lower concentrations of MCh as Kcna1-/- mice approached SD. MCh-induced seizures experienced by a subset of younger Kcna1-/- mice triggered death. Respiratory parameters of these younger Kcna1-/- mice resembled older near-SD Kcna1-/- mice. Kcna1 gene and protein were not expressed in Kcna1+/+ and Kcna1+/- lungs, and MCh-mediated airway smooth muscle contractions exhibited similar half-maximal effective concentration( EC50 ) in isolated Kcna1+/+ and Kcna1-/- trachea. SIGNIFICANCE: The Kcna1-/- model of SUDEP exhibits progressive respiratory dysfunction, which suggests a potential increased susceptibility for respiratory failure during severe seizures that may result in sudden death.


Asunto(s)
Apnea/genética , Muerte Súbita , Epilepsia del Lóbulo Temporal/fisiopatología , Hipoxia/genética , Canal de Potasio Kv.1.1/genética , Insuficiencia Respiratoria/genética , Animales , Apnea/complicaciones , Apnea/metabolismo , Broncoconstrictores/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epilepsia , Epilepsia del Lóbulo Temporal/complicaciones , Expresión Génica , Hiperventilación/inducido químicamente , Hipoxia/complicaciones , Hipoxia/metabolismo , Canal de Potasio Kv.1.1/metabolismo , Cloruro de Metacolina/farmacología , Ratones , Ratones Noqueados , Músculo Liso/efectos de los fármacos , Insuficiencia Respiratoria/complicaciones , Insuficiencia Respiratoria/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Taquipnea/complicaciones , Taquipnea/genética , Taquipnea/metabolismo , Volumen de Ventilación Pulmonar , Tráquea/efectos de los fármacos
6.
Epilepsia ; 58(8): 1440-1450, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28555877

RESUMEN

OBJECTIVE: We have previously found that the transcription factor peroxisome proliferator-activated receptor γ (PPARγ) contributes to the mechanism of action of the ketogenic diet (KD), an established treatment for pediatric refractory epilepsy. We have found that the KD increases brain PPARγ and that inhibition or genetic loss of PPARγ prevents the antiseizure effects of the KD on (1) acutely induced seizures in nonepileptic mice and (2) spontaneous recurrent seizures in epileptic mice. Here, we tested the hypothesis that adjuvant treatment of KD-treated mice with a PPARγ agonist, pioglitazone, would result in an additive effect. METHODS: Acute seizures were induced in three groups of C57Bl/6 mice by inhalation exposure to flurothyl gas. In Group 1, mice were weaned onto either a standard diet or KD comprised of a fat:carbohydrate/protein ratio of either 6:1, 3:1, or 1:1 for 2 weeks. In Group 2, vehicle or pioglitazone (0.1, 1, 10, 80 mg/kg) was administered 4 h prior to flurothyl exposure. In Group 3, vehicle or increasing doses of pioglitazone were administered to KD-treated mice 4 h prior to flurothyl exposure. Latency times to clonic seizures and generalized tonic-clonic (GTC) seizures were recorded, and isobolographic analysis was used to determine combinatorial interactions. RESULTS: Neither KD treatment nor pioglitazone alone or in combination affected clonic seizures. However, the latency to GTC seizures was dose-dependently and significantly increased by both KD (~57%, p < 0.05) and pioglitazone (~28%, p < 0.05). Coadministration of an ineffective 1:1 KD and pioglitazone resulted in ~47-55% (p < 0.05) increase in latency to GTC. Isobolographic analysis indicated a synergistic interaction of the KD and pioglitazone. SIGNIFICANCE: These results suggest coadministration may enable reduction of the KD ratio without loss of seizure protection. Such adjuvant treatment could improve quality of life and limit adverse effects of a classic KD or high-dose pioglitazone.


Asunto(s)
Dieta Cetogénica/métodos , Hipoglucemiantes/uso terapéutico , Convulsiones/dietoterapia , Convulsiones/tratamiento farmacológico , Tiazolidinedionas/uso terapéutico , Ácido 3-Hidroxibutírico/sangre , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Convulsivantes , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Flurotilo/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Pioglitazona , Tiempo de Reacción/efectos de los fármacos , Convulsiones/inducido químicamente
7.
Epilepsia ; 57(8): e178-82, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27346881

RESUMEN

Individuals with poorly controlled epilepsy have a higher risk for sudden unexpected death in epilepsy (SUDEP). With approximately one third of people with epilepsy not achieving adequate seizure control with current antiseizure drugs, there is a critical need to identify treatments that reduce risk factors for SUDEP. The Kcna1-null mutant mouse models risk factors and terminal events associated with SUDEP. In the current study, we demonstrate the progressive nature of epilepsy and sudden death in this model (mean age of mortality (± SEM), postnatal day [P] 42.8 ± 1.3) and tested the hypothesis that long-term treatment with the ketogenic diet (KD) will prolong the life of Kcna1-null mice. We found that the KD postpones disease progression by delaying the onset of severe seizures and increases the lifespan of these mutant mice by 47%. Future studies are needed to determine the mechanisms underlying the KD effects on longevity.


Asunto(s)
Muerte Súbita/etiología , Dieta Cetogénica/métodos , Epilepsia/complicaciones , Epilepsia/genética , Canal de Potasio Kv.1.1/deficiencia , Longevidad/efectos de los fármacos , Ácido 3-Hidroxibutírico/sangre , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Glucemia , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Electroencefalografía , Canal de Potasio Kv.1.1/genética , Longevidad/genética , Ratones , Ratones Transgénicos
8.
Epilepsia ; 55(5): e44-e49, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24702645

RESUMEN

The ketogenic diet (KD) is an effective therapy for pediatric refractory epilepsies; however, whether the KD changes the pathologic network oscillations generated by an epileptic brain remains unknown. We have reported that hippocampal CA3 regions of epileptic Kv1.1α knockout (KO) mice generate pathologic sharp waves (SPWs) and high-frequency oscillations (HFOs) that have higher incidence, longer duration, and fast ripples compared to wild-type (WT). Synaptic activity of hyperexcitable KO mossy fibers significantly decreased CA3 principal cell spike-timing reliability, which contributed to this network pathology. In addition, we have demonstrated that the KD reduces seizures by 75% in KO mice. Here, we determined whether 10- to 14-day in vivo KD treatment exerts disease-modifying effects that alter the spontaneous SPW-HFO complexes generated by the hippocampal CA3 region of KO mice in vitro using extracellular multielectrode array recordings. We found that KD treatment significantly attenuated the pathologic features of KO SPWs and ripples and reduced the incidence of fast ripples. The KD also improved spike-timing reliability of KO CA3 principal cells, decreased mossy fiber excitability, increased mossy fiber-CA3 paired-pulse ratios, and reduced coupling of field excitatory postsynaptic potentials and population spikes in the CA3 region. Collectively, these data indicate that KD treatment modulates CA3-generated pathologic oscillations by dampening hyperactive mossy fiber synapses.


Asunto(s)
Región CA3 Hipocampal/fisiopatología , Dieta Cetogénica , Electroencefalografía , Epilepsia/fisiopatología , Canal de Potasio Kv.1.1/genética , Animales , Epilepsia/dietoterapia , Epilepsia/genética , Potenciales Evocados/fisiología , Femenino , Expresión Génica/genética , Heterocigoto , Canal de Potasio Kv.1.1/fisiología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Fibras Musgosas del Hipocampo/fisiología , Sinapsis/fisiología , Técnicas de Cultivo de Tejidos
9.
Epilepsy Res ; 199: 107259, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38086218

RESUMEN

OBJECTIVE: Preclinical data report within subject modifiable ailments emerge weeks prior to SUDEP, including sleep disorders and cardiorespiratory changes; findings which support anecdotal clinical data. Here, we bridge preclinical findings with future clinical/preclinical studies, and survey whether caretakers or family members of victims noticed transient changes prior to SUDEP. The aim of this pilot study is to identify potential modifiable changes that may synergistically increase SUDEP risk for future research. METHODS: A mobile electronic survey was posted on SUDEP community websites. The survey queried whether changes in seizures, sleep, physical well-being, emotional well-being, cognition, breathing, or heart rate were noticed before SUDEP. RESULTS: The most profound finding was that 85% of victims had multiple transient ailments prior to SUDEP. Changes in seizures (28/54), and sleep (30/58) occurred in more than 50% of the victims and represent the most influential changes identified. The second and third most influential changes were a reduction in physical well-being (25/57) and emotional well-being (26/56). Changes were observed within the last two months of life in approximately one third of the cases, and more than four months prior to SUDEP in approximately one third of cases, indicating a potential time frame for proactive preventative strategies. Respondents also noted changes in cognition (16/55), breathing (9/54) or heart rate (8/55). Data indicate these changes may be associated with increased SUDEP risk within subject. Study limitations include the responses were based on memory, there was a potential for data to be over reported, and caretakers were not prompted to observe changes a priori, thus some existing changes may have gone unnoticed. SIGNIFICANCE: Data support the preclinical findings that transient, subclinical (i.e., not severe enough to require medical intervention), modifiable ailments may increase risk of SUDEP. This suggests that just as an epilepsy type can change over a lifetime and epilepsy type-specific treatments can reduce SUDEP risk, further personalization of SUDEP risk will improve our understanding as to whether variables contribute to risk differently across lifespan. Thus, with a dynamic capacity to change, differing factors may contribute to the distribution of risk probability within an individual at any given time. Understanding whether different combinations of transient changes are specific to epilepsy type, age, or sex needs to be determined to move the field forward in hopes of developing a personalized approach to preventative strategies.


Asunto(s)
Epilepsia , Muerte Súbita e Inesperada en la Epilepsia , Humanos , Proyectos Piloto , Muerte Súbita/epidemiología , Muerte Súbita/etiología , Convulsiones/epidemiología , Convulsiones/complicaciones , Encuestas y Cuestionarios , Factores de Riesgo
10.
Ecol Evol ; 13(11): e10664, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37933324

RESUMEN

Due to historical under-sampling of the deep ocean, the distributional ranges of mesopelagic zooplankton are not well documented, leading to uncertainty about the mechanisms that shape midwater zooplankton community composition. Using a combination of DNA metabarcoding (18S-V4 and mtCOI) and trait-based analysis, we characterized zooplankton diversity and community composition in the upper 1000 m of the northeast Pacific Ocean. We tested whether the North Pacific Transition Zone is a biogeographic boundary region for mesopelagic zooplankton. We also tested whether zooplankton taxa occupying different vertical habitats and exhibiting different ecological traits differed in the ranges of temperature, Chl-a, and dissolved oxygen conditions inhabited. The depth of the maximum taxonomic richness deepened with increasing latitude in the North Pacific. Community similarity in the mesopelagic zone also increased in comparison with the epipelagic zone, and no evidence was found for a biogeographic boundary between previously delineated mesopelagic biogeochemical provinces. Epipelagic zooplankton exhibited broader temperature and Chl-a ranges than mesopelagic taxa. Within the epipelagic, taxa with broader temperature and Chl-a ranges also had broader distributional ranges. However, mesopelagic taxa were distributed across wider dissolved oxygen ranges, and within the mesopelagic, only oxygen ranges covaried with distributional ranges. Environmental and distributional ranges also varied among traits, both for epipelagic taxa and mesopelagic taxa. The strongest differences in both environmental and distributional ranges were observed for taxa with or without diel vertical migration behavior. Our results suggest that species traits can influence the differential effects of physical dispersal and environmental selection in shaping biogeographic distributions.

11.
Nutrients ; 14(3)2022 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-35276972

RESUMEN

Ascorbic acid (AA; a.k.a. vitamin C) is well known for its cellular protection in environments of high oxidative stress. Even though physiological concentrations of AA in the brain are significant (0.2-10 mM), surprisingly little is known concerning the role of AA in synaptic neurotransmission under normal, non-disease state conditions. Here, we examined AA effects on neurotransmission, plasticity and spontaneous network activity (i.e., sharp waves and high frequency oscillations; SPW-HFOs), at the synapse between area 3 and 1 of the hippocampal cornu ammonis region (CA3 and CA1) using an extracellular multi-electrode array in in vitro mouse hippocampal slices. We found that AA decreased evoked field potentials (fEPSPs, IC50 = 0.64 mM) without affecting V50s or paired pulse facilitation indicating normal neurotransmitter release mechanisms. AA decreased presynaptic fiber volleys but did not change fiber volley-to-fEPSP coupling, suggesting reduced fEPSPs resulted from decreased fiber volleys. Inhibitory effects were also observed in CA1 stratum pyramidale where greater fEPSPs were required for population spikes in the presence of AA suggesting an impact on the intrinsic excitability of neurons. Other forms of synaptic plasticity and correlates of memory (i.e., short- and long-term potentiation) were also significantly reduced by AA as was the incidence of spontaneous SPW-HFOs. AA decreased SPW amplitude with a similar IC50 as fEPSPs (0.65 mM). Overall, these results indicate that under normal conditions AA significantly regulates neurotransmission, plasticity, and network activity by limiting excitability. Thus, AA may participate in refinement of signal processing and memory formation, as well as protecting against pathologic excitability.


Asunto(s)
Ácido Ascórbico , Transmisión Sináptica , Animales , Ácido Ascórbico/farmacología , Hipocampo , Potenciación a Largo Plazo/fisiología , Ratones , Plasticidad Neuronal/fisiología , Transmisión Sináptica/fisiología
12.
Brain Sci ; 11(6)2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203601

RESUMEN

Higher therapeutic concentrations of the antiseizure medication carbamazepine (CBZ) are associated with cognitive side effects. Hippocampal sharp wave-ripple complexes (SPW-Rs) are proposed to participate in memory consolidation during periods of quiet and slow-wave sleep. SPW-Rs are generated in the CA3 region and are regulated by multiple synaptic inputs. Here, we used a multi-electrode array to determine the effects of CBZ on SPW-Rs and synaptic transmission at multiple hippocampal synapses. Our results demonstrate that CBZ reduced SPW-Rs at therapeutically relevant concentrations (IC50 = 37 µM) and altered the core characteristics of ripples, important for information processing and consolidation. Moreover, CBZ inhibited neurotransmission in a synapse-specific manner. CBZ inhibition was most potent at the medial-perforant-path-to-CA3 and mossy-fiber-to-CA3 synapses (IC50s ~ 30 and 60 µM, respectively) and least potent at medial-perforant-path-to-dentate granule cell synapses (IC50 ~ 120 µM). These results suggest that the synapse-specific CBZ inhibition of neurotransmission reduces SPW-Rs and that the CBZ inhibition of SPW-Rs may underlie the cognitive impairments observed with therapeutic doses of CBZ.

13.
Eur J Pharmacol ; 913: 174656, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34838797

RESUMEN

Drug resistant epilepsy affects ∼30% of people with epilepsy and is associated with epilepsy syndromes with frequent and multiple types of seizures, lesions or cytoarchitectural abnormalities, increased risk of mortality and comorbidities such as cognitive impairment and sleep disorders. A limitation of current preclinical models is that spontaneous seizures with comorbidities take time to induce and test, thus making them low-throughput. Kcna1-null mice exhibit all the characteristics of drug resistant epilepsy with spontaneous seizures and comorbidities occurring naturally; thus, we aimed to determine whether they also demonstrate pharmacoresistanct seizures and the impact of medications on their sleep disorder comorbidity. In this exploratory study, Kcna1-null mice were treated with one of four conventional antiseizure medications, carbamazepine, levetiracetam, phenytoin, and phenobarbital using a moderate throughput protocol (vehicle for 2 days followed by 2 days of treatment with high therapeutic doses selected based on published data in the 6 Hz model of pharmacoresistant seizures). Spontaneous recurrent seizures and vigilance states were recorded with video-EEG/EMG. Carbamazepine, levetiracetam and phenytoin had partial efficacy (67%, 75% and 33% were seizure free, respectively), whereas phenobarbital was fully efficacious and conferred seizure freedom to all mice. Thus, seizures of Kcna1-null mice appear to be resistant to three of the drugs tested. Levetiracetam failed to affect sleep architecture, carbamazepine and phenytoin had moderate effects, and phenobarbital, as predicted, restored sleep architecture. Data suggest Kcna1-null mice may be a moderate throughput model of drug resistant epilepsy useful in determining mechanisms of pharmacoresistance and testing novel therapeutic strategies.


Asunto(s)
Anticonvulsivantes/farmacología , Epilepsia Refractaria/tratamiento farmacológico , Canal de Potasio Kv.1.1/genética , Convulsiones/tratamiento farmacológico , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Animales , Anticonvulsivantes/uso terapéutico , Modelos Animales de Enfermedad , Epilepsia Refractaria/complicaciones , Epilepsia Refractaria/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Recurrencia , Convulsiones/genética , Trastornos del Sueño-Vigilia/complicaciones , Trastornos del Sueño-Vigilia/genética
15.
Epilepsy Res ; 147: 71-74, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30261354

RESUMEN

We have previously found that the transcription factor PPARγ2 contributes to the mechanism of action of the ketogenic diet (KD), an established treatment for pediatric refractory epilepsy. Among the wide-array of genes regulated by PPARγ, previous studies have suggested that antioxidants such as catalase may have prominent roles in KD neuroprotective and antiseizure effects. Here, we tested the hypothesis that the KD increases catalase through activation of PPARγ2, and that this action is part of the mechanism of antiseizure efficacy of the KD. We determined catalase mRNA and protein expression in hippocampal tissue from epileptic Kcna1-/- mice, Pparγ2+/+ mice and Pparγ2-/- mice. We found that a KD increases hippocampal catalase expression in Kcna1-/- and Pparγ2+/+ mice, but not Pparγ2-/- mice. Next, we determined whether catalase contributes to KD seizure protection. We found that the KD reduces pentylenetetrazole (PTZ)-induced seizures; however, pretreatment with a catalase inhibitor occluded KD effects on PTZ seizures. These results suggest that the KD regulates catalase expression through PPARγ2 activation, and that catalase may contribute to the KD antiseizure efficacy.


Asunto(s)
Catalasa/metabolismo , Dieta Cetogénica , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , PPAR gamma/metabolismo , Convulsiones/patología , Animales , Catalasa/genética , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/genética , Hipocampo/efectos de los fármacos , Canal de Potasio Kv.1.1/deficiencia , Canal de Potasio Kv.1.1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , PPAR gamma/genética , Pentilenotetrazol/toxicidad , ARN Mensajero/metabolismo , Convulsiones/inducido químicamente , Convulsiones/dietoterapia , Azida Sódica/farmacología
16.
Neuropsychopharmacology ; 32(3): 646-57, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16738542

RESUMEN

The hypothesis that prior cannabis exposure increases the likelihood of becoming addicted to other drugs can be evaluated by giving rats a history of tetrahydrocannabinol (THC) exposure, then allowing them to self-administer other drugs. In Experiment 1, THC pre-exposure did not alter the acquisition of cocaine self-administration or the amount of cocaine taken under a fixed-ratio 1 (FR1) schedule, with one response required for each injection. Under a progressive-ratio schedule, with the response requirement increasing exponentially with each injection, cocaine-seeking was significantly reduced in THC-exposed rats, suggesting that the regimen of THC exposure used in the present study caused cocaine to be devalued as a reinforcer. In contrast, in an earlier study that used the same regimen, a history of THC exposure did not alter the value of heroin as a reinforcer under the progressive-ratio schedule, but it increased heroin self-administration under the FR1 schedule. Experiment 2 examined how this regimen of THC pre-exposure alters the locomotor effects of cocaine and heroin. THC pre-exposure produced cross-tolerance to the motor-depressant effects of heroin; this may explain the shortened post-injection pauses exhibited by THC-exposed rats under FR1 heroin self-administration. When given cocaine, THC-exposed rats exhibited normal increases in locomotion, but they avoided the center of the open field, suggesting that this THC pre-exposure regimen enhances the anxiogenic effects of cocaine. This enhanced anxiogenic effect-which was verified in Experiment 3 using another model of anxiety, the light-dark test-may explain the reduced reinforcing value of cocaine observed in THC-exposed rats in Experiment 1.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Ansiedad/inducido químicamente , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Dronabinol/farmacología , Refuerzo en Psicología , Animales , Ansiedad/fisiopatología , Conducta Animal , Conducta de Elección/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Interacciones Farmacológicas , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Esquema de Refuerzo , Autoadministración
17.
Exp Neurol ; 287(Pt 1): 54-64, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27527983

RESUMEN

The ketogenic diet (KD) is an effective therapy primarily used in pediatric patients whom are refractory to current anti-seizure medications. The mechanism of the KD is not completely understood, but is thought to involve anti-inflammatory and anti-oxidant processes. The nutritionally-regulated transcription factor peroxisome proliferator activated receptor gamma, PPARγ, regulates genes involved in anti-inflammatory and anti-oxidant pathways. Moreover, endogenous ligands of PPARγ include fatty acids suggesting a potential role in the effects of the KD. Here, we tested the hypothesis that PPARγ contributes to the anti-seizure efficacy of the KD. We found that the KD increased nuclear protein content of the PPARγ2 splice variant by 2-4 fold (P<0.05) in brain homogenates from wild-type (WT) and epileptic Kv1.1 knockout (KO) mice, while not affecting PPARγ1. The KD reduced the frequency of seizures in Kv1.1KO mice by ~70% (P<0.01). GW9662, a PPARγ antagonist, prevented KD-mediated changes in PPARγ2 expression and prevented the anti-seizure efficacy of the KD in Kv1.1KO mice. Further supporting the association of PPARγ2 in mediating KD actions, the KD significantly prolonged the latency to flurothyl-induced seizure in WT mice by ~20-35% (P<0.01), but was ineffective in PPARγ2KO mice and neuron-specific PPARγKO mice. Finally, administering the PPARγ agonist pioglitazone increased PPARγ2 expression by 2-fold (P<0.01) and reduced seizures in Kv1.1KO mice by ~80% (P<0.01). Our findings implicate brain PPARγ2 among the mechanisms by which the KD reduces seizures and strongly support the development of PPARγ2 as a therapeutic target for severe, refractory epilepsy.


Asunto(s)
Encéfalo/metabolismo , Dieta Cetogénica/métodos , Epilepsia/dietoterapia , Epilepsia/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , PPAR gamma/metabolismo , Ácido 3-Hidroxibutírico/sangre , Factores de Edad , Anilidas/farmacología , Anilidas/uso terapéutico , Animales , Animales Recién Nacidos , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Glucemia/efectos de los fármacos , Glucemia/genética , Encéfalo/efectos de los fármacos , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Líquidos/fisiología , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Flurotilo/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Hipoglucemiantes/farmacología , Canal de Potasio Kv.1.1/deficiencia , Canal de Potasio Kv.1.1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/genética , Pioglitazona , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico
18.
Sleep ; 39(2): 357-68, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26446112

RESUMEN

STUDY OBJECTIVE: Comorbid sleep disorders occur in approximately one-third of people with epilepsy. Seizures and sleep disorders have an interdependent relationship where the occurrence of one can exacerbate the other. Orexin, a wake-promoting neuropeptide, is associated with sleep disorder symptoms. Here, we tested the hypothesis that orexin dysregulation plays a role in the comorbid sleep disorder symptoms in the Kcna1-null mouse model of temporal lobe epilepsy. METHODS: Rest-activity was assessed using infrared beam actigraphy. Sleep architecture and seizures were assessed using continuous video-electroencephalography-electromyography recordings in Kcna1-null mice treated with vehicle or the dual orexin receptor antagonist, almorexant (100 mg/kg, intraperitoneally). Orexin levels in the lateral hypothalamus/perifornical region (LH/P) and hypothalamic pathology were assessed with immunohistochemistry and oxygen polarography. RESULTS: Kcna1-null mice have increased latency to rapid eye movement (REM) sleep onset, sleep fragmentation, and number of wake epochs. The numbers of REM and non-REM (NREM) sleep epochs are significantly reduced in Kcna1-null mice. Severe seizures propagate to the wake-promoting LH/P where injury is apparent (indicated by astrogliosis, blood-brain barrier permeability, and impaired mitochondrial function). The number of orexin-positive neurons is increased in the LH/P compared to wild-type LH/P. Treatment with a dual orexin receptor antagonist significantly increases the number and duration of NREM sleep epochs and reduces the latency to REM sleep onset. Further, almorexant treatment reduces the incidence of severe seizures and overall seizure burden. Interestingly, we report a significant positive correlation between latency to REM onset and seizure burden in Kcna1-null mice. CONCLUSION: Dual orexin receptor antagonists may be an effective sleeping aid in epilepsy, and warrants further study on their somnogenic and ant-seizure effects in other epilepsy models.


Asunto(s)
Canal de Potasio Kv.1.1/deficiencia , Canal de Potasio Kv.1.1/genética , Antagonistas de los Receptores de Orexina/farmacología , Receptores de Orexina/metabolismo , Convulsiones/prevención & control , Sueño/efectos de los fármacos , Sueño/fisiología , Acetamidas/farmacología , Acetamidas/uso terapéutico , Actigrafía , Animales , Electroencefalografía , Electromiografía , Epilepsia del Lóbulo Temporal/complicaciones , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/fisiopatología , Área Hipotalámica Lateral/citología , Área Hipotalámica Lateral/fisiología , Isoquinolinas/farmacología , Isoquinolinas/uso terapéutico , Ratones , Ratones Noqueados , Neuronas/fisiología , Antagonistas de los Receptores de Orexina/uso terapéutico , Orexinas/metabolismo , Oxígeno/metabolismo , Descanso/fisiología , Convulsiones/complicaciones , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología , Privación de Sueño/complicaciones , Privación de Sueño/tratamiento farmacológico , Privación de Sueño/fisiopatología , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Vigilia/efectos de los fármacos , Vigilia/fisiología
19.
Neuropsychopharmacology ; 30(11): 2046-57, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15870833

RESUMEN

Accumulating evidence suggests that the endogenous cannabinoid system is involved in the reinforcing effects of heroin. In rats intravenously self-administering heroin, we investigated effects of cannabinoid CB1 receptor agonists and compounds that block transport or metabolism of the endogenous cannabinoid anandamide. The natural cannnabinoid CB1 receptor agonist delta-9-tetrahydrocannabinol (THC, 0.3-3 mg/kg i.p.) did not alter self-administration of heroin under a fixed-ratio one (FR1) schedule, except at a high 3 mg/kg dose which decreased heroin self-administration. Under a progressive-ratio schedule, however, THC dose-dependently increased the number of 50 mug/kg heroin injections self-administered per session and the maximal ratio completed (break-point), with peak increases at 1 mg/kg THC. In addition, 1 mg/kg THC increased break-points and injections self-administered over a wide range of heroin injection doses (25-100 microg/kg), indicating an increase in heroin's reinforcing efficacy and not its potency. The synthetic cannabinoid CB1 receptor agonist WIN55,212-2 (0.3-3 mg/kg i.p.) had effects similar to THC under the progressive-ratio schedule. In contrast, AM-404 (1-10 mg/kg i.p.), an inhibitor of transport of anandamide, and URB-597 (0.01-0.3 mg/kg i.p.), an inhibitor of the enzyme fatty acid amide hydrolase (FAAH) that degrades anandamide, or their combination, did not increase reinforcing efficacy of heroin at any dose tested. Thus, activation of cannabinoid CB1 receptors facilitates the reinforcing efficacy of heroin and this appears to be mediated by interactions between cannabinoid CB1 receptors and mu-opioid receptors and their signaling pathways, rather than by an opioid-induced release of endogenous cannabinoids.


Asunto(s)
Moduladores de Receptores de Cannabinoides/antagonistas & inhibidores , Cannabinoides/agonistas , Condicionamiento Operante/efectos de los fármacos , Heroína/administración & dosificación , Narcóticos/administración & dosificación , Refuerzo en Psicología , Animales , Ácidos Araquidónicos/farmacología , Conducta Animal , Benzamidas/farmacología , Benzoxazinas , Moduladores de Receptores de Cannabinoides/agonistas , Cannabinoides/antagonistas & inhibidores , Carbamatos/farmacología , Relación Dosis-Respuesta a Droga , Dronabinol/farmacología , Interacciones Farmacológicas , Masculino , Morfolinas/farmacología , Naftalenos/farmacología , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Esquema de Refuerzo , Autoadministración/psicología , Factores de Tiempo
20.
Exp Neurol ; 251: 84-90, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24270080

RESUMEN

Mitochondria actively participate in neurotransmission by providing energy (ATP) and maintaining normative concentrations of reactive oxygen species (ROS) in both presynaptic and postsynaptic elements. In human and animal epilepsies, ATP-producing respiratory rates driven by mitochondrial respiratory complex (MRC) I are reduced, antioxidant systems are attenuated and oxidative damage is increased. We report that MRCI-driven respiration and functional uncoupling (an inducible antioxidant mechanism) are reduced and levels of H2O2 are elevated in mitochondria isolated from KO mice. Experimental impairment of MRCI in WT hippocampal slices via rotenone reduces paired-pulse ratios (PPRs) at mossy fiber-CA3 synapses (resembling KO PPRs), and exacerbates seizure-like events in vitro. Daily treatment with AATP [a combination therapy composed of ascorbic acid (AA), alpha-tocopherol (T), sodium pyruvate (P) designed to synergistically target mitochondrial impairments] improved mitochondrial functions, mossy fiber PPRs, and reduced seizure burden index (SBI) scores and seizure incidence in KO mice. AATP pretreatment reduced severity of KA-induced seizures resulting in 100% protection from the severe tonic-clonic seizures in WT mice. These data suggest that restoration of bioenergetic homeostasis in the brain may represent a viable anti-seizure target for temporal lobe epilepsy.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/prevención & control , Adenosina Trifosfato/administración & dosificación , Adenosina Trifosfato/análogos & derivados , Animales , Ácido Ascórbico/uso terapéutico , Modelos Animales de Enfermedad , Estimulación Eléctrica , Electroencefalografía , Epilepsia del Lóbulo Temporal/genética , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/fisiopatología , Peróxido de Hidrógeno/metabolismo , Técnicas In Vitro , Ácido Kaínico/toxicidad , Canal de Potasio Kv.1.1/deficiencia , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ácido Pirúvico/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Respiración/efectos de los fármacos , Respiración/genética , Convulsiones/inducido químicamente , Convulsiones/prevención & control , Convulsiones/terapia , alfa-Tocoferol/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA