Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-39000060

RESUMEN

Neurodegenerative diseases (NDs) represent an unsolved problem to date with an ever-increasing population incidence. Particularly, Alzheimer's disease (AD) is the most widespread ND characterized by an accumulation of amyloid aggregates of beta-amyloid (Aß) and Tau proteins that lead to neuronal death and subsequent cognitive decline. Although neuroimaging techniques are needed to diagnose AD, the investigation of biomarkers within body fluids could provide important information on neurodegeneration. Indeed, as there is no definitive solution for AD, the monitoring of these biomarkers is of strategic importance as they are useful for both diagnosing AD and assessing the progression of the neurodegenerative state. In this context, exercise is known to be an effective non-pharmacological management strategy for AD that can counteract cognitive decline and neurodegeneration. However, investigation of the concentration of fluid biomarkers in AD patients undergoing exercise protocols has led to unclear and often conflicting results, suggesting the need to clarify the role of exercise in modulating fluid biomarkers in AD. Therefore, this critical literature review aims to gather evidence on the main fluid biomarkers of AD and the modulatory effects of exercise to clarify the efficacy and usefulness of this non-pharmacological strategy in counteracting neurodegeneration in AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Biomarcadores , Ejercicio Físico , Proteínas tau , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Humanos , Biomarcadores/metabolismo , Ejercicio Físico/fisiología , Péptidos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Terapia por Ejercicio/métodos
2.
Int J Mol Sci ; 21(3)2020 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-31979108

RESUMEN

γ-Aminobutyric acid type A receptors (GABAARs) are the main inhibitory mediators in the central nervous system (CNS). GABAARs are pentameric ligand gated ion channels, and the main subunit composition is usually 2α2ßγ, with various isotypes assembled within a set of 19 different subunits. The inhibitory function is mediated by chloride ion movement across the GABAARs, activated by synaptic GABA release, reducing neuronal excitability in the adult CNS. Several studies highlighted the importance of GABA-mediated transmission during neuro-development, and its involvement in different neurological and neurodevelopmental diseases, from anxiety to epilepsy. However, while it is well known how different classes of drugs are able to modulate the GABAARs function (benzodiazepines, barbiturates, neurosteroids, alcohol), up to now little is known about GABAARs and cannabinoids interaction in the CNS. Endocannabinoids and phytocannabinoids are lately emerging as a new class of promising drugs for a wide range of neurological conditions, but their safety as medication, and their mechanisms of action are still to be fully elucidated. In this review, we will focus our attention on two of the most promising molecules (Δ9-tetrahydrocannabinol; Δ9-THC and cannabidiol; CBD) of this new class of drugs and their possible mechanism of action on GABAARs.


Asunto(s)
Cannabinoides/farmacología , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Receptores de GABA-A/metabolismo , Animales , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Humanos
3.
Proc Natl Acad Sci U S A ; 113(11): 3060-5, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26929355

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting motor neurons that leads to progressive paralysis of skeletal muscle. Studies of ALS have revealed defects in expression of acetylcholine receptors (AChRs) in skeletal muscle that occur even in the absence of motor neuron anomalies. The endocannabinoid palmitoylethanolamide (PEA) modified the clinical conditions in one ALS patient, improving muscle force and respiratory efficacy. By microtransplanting muscle membranes from selected ALS patients into Xenopus oocytes, we show that PEA reduces the desensitization of acetylcholine-evoked currents after repetitive neurotransmitter application (i.e., rundown). The same effect was observed using muscle samples from denervated (non-ALS) control patients. The expression of human recombinant α1ß1γδ (γ-AChRs) and α1ß1εδ AChRs (ε-AChRs) in Xenopus oocytes revealed that PEA selectively affected the rundown of ACh currents in ε-AChRs. A clear up-regulation of the α1 subunit in muscle from ALS patients compared with that from non-ALS patients was found by quantitative PCR, but no differential expression was found for other subunits. Clinically, ALS patients treated with PEA showed a lower decrease in their forced vital capacity (FVC) over time as compared with untreated ALS patients, suggesting that PEA can enhance pulmonary function in ALS. In the present work, data were collected from a cohort of 76 ALS patients and 17 denervated patients. Our results strengthen the evidence for the role of skeletal muscle in ALS pathogenesis and pave the way for the development of new drugs to hamper the clinical effects of the disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Etanolaminas/uso terapéutico , Terapia Molecular Dirigida , Músculo Esquelético/efectos de los fármacos , Ácidos Palmíticos/uso terapéutico , Receptores Nicotínicos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Amidas , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/trasplante , Etanolaminas/farmacología , Femenino , Humanos , Masculino , Microinyecciones , Persona de Mediana Edad , Desnervación Muscular , Músculo Esquelético/ultraestructura , Unión Neuromuscular/fisiopatología , Oocitos , Ácidos Palmíticos/farmacología , Receptores Nicotínicos/fisiología , Proteínas Recombinantes de Fusión/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Método Simple Ciego , Xenopus laevis
4.
Neurobiol Dis ; 115: 59-68, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29621596

RESUMEN

Epilepsy is one of the most common chronic neurological diseases, and its pharmacological treatment holds great importance for both physicians and national authorities, especially considering the high proportion of drug-resistant patients (about 30%). Lacosamide (LCM) is an effective and well-tolerated new-generation antiepileptic drug (AED), currently licensed as add-on therapy for partial-onset seizures. However, LCM mechanism of action is still a matter of debate, although its effect on the voltage sensitive sodium channels is by far the most recognized. This study aimed to retrospectively analyze a cohort of 157 drug-resistant patients treated with LCM to describe the most common and effective therapeutic combinations and to investigate if the LCM can affect also GABAA-mediated neurotransmission as previously shown for levetiracetam (LEV). In our cohort, LEV resulted the compound most frequently associated with LCM in the responder subgroup. We therefore translated this clinical observation into the laboratory bench by taking advantage of the technique of "membrane micro-transplantation" in Xenopus oocytes and electrophysiological approaches to study human GABAA-evoked currents. In cortical brain tissues from refractory epileptic patients, we found that LCM reduces the use-dependent GABA impairment (i.e., "rundown") that it is considered one of the specific hallmarks of drug-resistant epilepsies. Notably, in line with our clinical observations, we found that the co-treatment with subthreshold concentrations of LCM and LEV, which had no effect on GABAA currents on their own, reduced GABA impairment in drug-resistant epileptic patients, and this effect was blocked by PKC inhibitors. Our findings demonstrate, for the first time, that LCM targets GABAA receptors and that it can act synergistically with LEV, improving the GABAergic function. This novel mechanism might contribute to explain the clinical efficacy of LCM-LEV combination in several refractory epileptic patients.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Epilepsia Refractaria/tratamiento farmacológico , Lacosamida/administración & dosificación , Levetiracetam/administración & dosificación , Receptores de GABA-A/fisiología , Adulto , Anciano , Animales , Anticonvulsivantes/sangre , Estudios de Cohortes , Epilepsia Refractaria/sangre , Epilepsia Refractaria/diagnóstico , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Humanos , Lacosamida/sangre , Levetiracetam/sangre , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento , Xenopus , Adulto Joven
5.
Epilepsia ; 59(11): 2106-2117, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30306542

RESUMEN

OBJECTIVE: Dravet syndrome is a rare neurodevelopmental disease, characterized by general cognitive impairment and severe refractory seizures. The majority of patients carry the gene mutation SCN1A, leading to a defective sodium channel that contributes to pathogenic brain excitability. A γ-aminobutyric acid (GABAergic) impairment, as in other neurodevelopmental diseases, has been proposed as an additional mechanism, suggesting that seizures could be alleviated by GABAergic therapies. However, up to now the physiological mechanisms underlying the GABAergic dysfunction in Dravet syndrome are still unknown due to the scarce availability of this brain tissue. Here we studied, for the first time, human GABAA -evoked currents using cortical brain tissue from Dravet syndrome patients. METHODS: We transplanted in Xenopus oocytes cell membranes obtained from brain tissues of autopsies of Dravet syndrome patients, tuberous sclerosis complex patients as a pathological comparison, and age-matched controls. Additionally, experiments were performed on oocytes expressing human α1ß2γ2 and α1ß2 GABAA receptors. GABAA currents were recorded using the two-microelectrodes voltage-clamp technique. Quantitative real-time polymerase chain reaction, immunohistochemistry, and double-labeling techniques were carried out on the same tissue samples. RESULTS: We found (1) a decrease in GABA sensitivity in Dravet syndrome compared to controls, which was related to an increase in α4- relative to α1-containing GABAA receptors; (2) a shift of the GABA reversal potential toward more depolarizing values in Dravet syndrome, and a parallel increase of the chloride transporters NKCC1/KCC2 expression ratio; (3) an increase of GABAA currents induced by low doses of cannabidiol both in Dravet syndrome and tuberous sclerosis complex comparable to that induced by a classical benzodiazepine, flunitrazepam, that still persists in γ-less GABAA receptors. SIGNIFICANCE: Our study indicates that a dysfunction of the GABAergic system, considered as a feature of brain immaturity, together with defective sodium channels, can contribute to a general reduction of inhibitory efficacy in Dravet brain, suggesting that GABAA receptors could be a target for new therapies.


Asunto(s)
Corteza Cerebral/patología , Epilepsias Mioclónicas/patología , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Adulto , Animales , Anticonvulsivantes/farmacología , Cannabidiol/farmacología , Membrana Celular/trasplante , Corteza Cerebral/ultraestructura , Niño , Preescolar , Estimulación Eléctrica , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/metabolismo , Femenino , Humanos , Larva , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Persona de Mediana Edad , ARN Mensajero/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/genética , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Xenopus , Ácido gamma-Aminobutírico/farmacología
6.
Neurobiol Dis ; 95: 93-101, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27425893

RESUMEN

Tuberous sclerosis complex (TSC) is a rare multi-system genetic disease characterized by several neurological disorders, the most common of which is the refractory epilepsy caused by highly epileptogenic cortical lesions. Previous studies suggest an alteration of GABAergic and glutamatergic transmission in TSC brain indicating an unbalance of excitation/inhibition that can explain, at least in part, the high incidence of epilepsy in these patients. Here we investigate whether TSC cortical tissues could retain GABAA and AMPA receptors at early stages of human brain development thus contributing to the generation and recurrence of seizures. Given the limited availability of pediatric human brain specimens, we used the microtransplantation method of injecting Xenopus oocytes with membranes from TSC cortical tubers and control brain tissues. Moreover, qPCR was performed to investigate the expression of GABAA and AMPA receptor subunits (GABAA α1-5, ß3, γ2, δ; GluA1, GluA2) and cation chloride co-transporters NKCC1 and KCC2. The evaluation of nine human cortical brain samples, from 15 gestation weeks to 15years old, showed a progressive shift towards more hyperpolarized GABAA reversal potential (EGABA). This shift was associated with a differential expression of the chloride cotransporters NKCC1 and KCC2. Furthermore, the GluA1/GluA2 mRNA ratio of expression paralleled the development process. On the contrary, in oocytes micro-transplanted with epileptic TSC tuber tissue from seven patients, neither the GABAA reversal potential nor the GluA1/GluA2 expression showed similar developmental changes. Our data indicate for the first time, that in the same cohort of TSC patients, the pattern of both GABAAR and GluA1/GluA2 functions retains features that are typical of an immature brain. These observations support the potential contribution of altered receptor function to the epileptic disorder of TSC and may suggest novel therapeutic approaches. Furthermore, our findings strengthen the novel hypothesis that other developmental brain diseases can share the same hallmarks of immaturity leading to intractable seizures.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Epilepsia/etiología , Esclerosis Tuberosa/patología , Esclerosis Tuberosa/fisiopatología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encefalopatías/patología , Niño , Estudios de Cohortes , Femenino , Humanos , Oocitos , Receptores de GABA-A/metabolismo , Convulsiones/fisiopatología , Simportadores/metabolismo , Esclerosis Tuberosa/genética , Xenopus
7.
Pharmacol Res ; 113(Pt A): 421-425, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27659220

RESUMEN

Mesial temporal lobe epilepsy (MTLE) is frequently associated with hippocampal sclerosis (Hs), possibly caused by a primary brain injury that occurs a long time before the appearance of neurological symptoms. MTLE-Hs is, however, a heterogeneous condition that evolves with time, involving both environmental and genetic components. Recent experimental studies emphasize that drugs or drug combinations that target modulation and circuitry reorganization of the epileptogenic networks favorably modify the complex molecular and cellular alterations underlying MTLE. In particular, the link between neuroinflammation, GABAAR and epilepsy has been extensively studied mainly because of the relevant therapeutic implications that the pharmacological modulation of these phenomena would have in the clinical practice. In this review, we briefly summarize the studies that could pave the road to develop new disease-modifying therapeutic strategies for pharmacoresistant MTLE patients. Both clinical observations in human MTLE and experimental findings will be discussed, highlighting the potential modulatory crosstalk between the deregulation of the inhibitory (GABAergic) transmission and the sustained activation of the innate immune response.


Asunto(s)
Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Preparaciones Farmacéuticas/administración & dosificación , Animales , GABAérgicos/uso terapéutico , Hipocampo/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos
8.
Neurobiol Dis ; 82: 311-320, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26168875

RESUMEN

Temporal lobe epilepsy (TLE) is the most prevalent form of adult focal onset epilepsy often associated with drug-resistant seizures. Numerous studies suggest that neuroinflammatory processes are pathologic hallmarks of both experimental and human epilepsy. In particular, the interleukin (IL)-1ß/IL-1 receptor type 1 (R1) axis is activated in epileptogenic tissue, where it contributes significantly to the generation and recurrence of seizures in animal models. In this study, we investigated whether IL-1ß affects the GABA-evoked currents (I(GABA)) in TLE tissue from humans. Given the limited availability of fresh human brain specimens, we used the "microtransplantation" method of injecting Xenopus oocytes with membranes from surgically resected hippocampal and cortical tissue from 21 patients with TLE and hippocampal sclerosis (HS), hippocampal tissue from five patients with TLE without HS, and autoptic and surgical brain specimens from 15 controls without epilepsy. We report the novel finding that pathophysiological concentrations of IL-1ß decreased the I(GABA) amplitude by up to 30% in specimens from patients with TLE with or without HS, but not in control tissues. This effect was reproduced by patch-clamp recordings on neurons in entorhinal cortex slices from rats with chronic epilepsy, and was not observed in control slices. In TLE specimens from humans, the IL-1ß effect was mediated by IL-1R1 and PKC. We also showed that IL-1R1 and IRAK1, the proximal kinase mediating the IL-1R1 signaling, are both up-regulated in the TLE compared with control specimens, thus supporting the idea that the IL-1ß/IL-R1 axis is activated in human epilepsy. Our findings suggest a novel mechanism possibly underlying the ictogenic action of IL-1ß, thus suggesting that this cytokine contributes to seizure generation in human TLE by reducing GABA-mediated neurotransmission.


Asunto(s)
Corteza Cerebral/fisiopatología , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/fisiopatología , Interleucina-1beta/metabolismo , Receptores de GABA-A/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Corteza Cerebral/patología , Corteza Cerebral/cirugía , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/patología , Epilepsia del Lóbulo Temporal/cirugía , Femenino , GABAérgicos/administración & dosificación , Hipocampo/patología , Hipocampo/cirugía , Humanos , Interleucina-1beta/administración & dosificación , Ácido Kaínico , Masculino , Persona de Mediana Edad , Oocitos , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Técnicas de Cultivo de Tejidos , Trasplante Heterólogo/métodos , Xenopus , Adulto Joven
9.
Membranes (Basel) ; 14(3)2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38535283

RESUMEN

Among the most prevalent neurological disorders, epilepsy affects about 1% of the population worldwide. We previously found, using human epileptic tissues, that GABAergic neurotransmission impairment is a key mechanism that drives the pathological phenomena that ultimately lead to generation and recurrence of seizures. Using both a "microtransplantation technique" and synaptosomes preparations from drug-resistant temporal lobe epilepsies (TLEs), we used the technique of two-electrode voltage clamp to record GABA-evoked currents, focusing selectively on the synaptic "fast inhibition" mediated by low-affinity GABAA receptors. Here, we report that the use-dependent GABA current desensitization (i.e., GABA rundown, which is evoked by applying to the cells consecutive pulses of GABA, at high concentration), which is a distinguishing mark of TLE, is mainly dependent on a dysfunction that affects synaptic GABAA receptors. In addition, using the same approaches, we recorded a depolarized GABA reversal potential in synaptosomes samples from the human epileptic subicula of TLE patients. These results, which confirm previous experiments using total membranes, suggest an altered chloride homeostasis in the synaptic area. Finally, the lack of a Zn2+ block of GABA-evoked currents using the synaptosomes supports the enrichment of "synaptic fast inhibitory" GABAA receptors in this preparation. Altogether, our findings suggest a pathophysiological role of low-affinity GABAA receptors at the synapse, especially during the fast and repetitive GABA release underlying recurrent seizures.

10.
Neurobiol Dis ; 55: 110-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23454193

RESUMEN

HIV-1 associated neurocognitive disorders (HAND) are a major complication of HIV-1 infection. The mechanism(s) underlying HAND are not completely understood but, based on in vitro studies, the HIV-1 Tat protein may play an important role. In this study, the effect of prolonged exposure to endogenously produced Tat in the brain was investigated using a tat-transgenic (TT) mouse model constitutively expressing the HIV-1 tat gene. We found that stimulus-evoked glutamate exocytosis in the hippocampus and cortex was significantly increased in TT as compared with wild-type control (CC) mice, while GABA exocytosis was unchanged in the hippocampus and decreased in the cortex. This suggests that Tat generates a latent hyper-excitability state, which favors the detrimental effects of neurotoxic and/or excitotoxic agents. To challenge this idea, TT mice were tested for susceptibility to kainate-induced seizures and neurodegeneration, and found to exhibit significantly greater responses to the convulsant agent than CC mice. These results support the concept that constitutive expression of tat in the brain generates a latent excitatory state, which may increase the negative effects of damaging insults. These events may play a key role in the development of HAND.


Asunto(s)
Encéfalo/patología , Enfermedades del Sistema Nervioso/patología , Enfermedades del Sistema Nervioso/virología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/virología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Productos del Gen tat/farmacología , Ácido Kaínico/toxicidad , Masculino , Ratones , Ratones Transgénicos , Neurotransmisores/metabolismo , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Estadísticas no Paramétricas , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
11.
Antioxidants (Basel) ; 12(1)2023 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-36670973

RESUMEN

Down syndrome (DS) is the most frequent genetic cause of intellectual disability and is strongly associated with Alzheimer's disease (AD). Brain insulin resistance greatly contributes to AD development in the general population and previous studies from our group showed an early accumulation of insulin resistance markers in DS brain, already in childhood, and even before AD onset. Here we tested the effects promoted in Ts2Cje mice by the intranasal administration of the KYCCSRK peptide known to foster insulin signaling activation by directly interacting and activating the insulin receptor (IR) and the AKT protein. Therefore, the KYCCSRK peptide might represent a promising molecule to overcome insulin resistance. Our results show that KYCCSRK rescued insulin signaling activation, increased mitochondrial complexes levels (OXPHOS) and reduced oxidative stress levels in the brain of Ts2Cje mice. Moreover, we uncovered novel characteristics of the KYCCSRK peptide, including its efficacy in reducing DYRK1A (triplicated in DS) and BACE1 protein levels, which resulted in reduced AD-like neuropathology in Ts2Cje mice. Finally, the peptide elicited neuroprotective effects by ameliorating synaptic plasticity mechanisms that are altered in DS due to the imbalance between inhibitory vs. excitatory currents. Overall, our results represent a step forward in searching for new molecules useful to reduce intellectual disability and counteract AD development in DS.

12.
Inflamm Regen ; 43(1): 19, 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36895050

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a heterogeneous disease in terms of onset and progression rate. This may account for therapeutic clinical trial failure. Transgenic SOD1G93A mice on C57 or 129Sv background have a slow and fast disease progression rate, mimicking the variability observed in patients. Based on evidence inferring the active influence of skeletal muscle on ALS pathogenesis, we explored whether dysregulation in hindlimb skeletal muscle reflects the phenotypic difference between the two mouse models. METHODS: Ex vivo immunohistochemical, biochemical, and biomolecular methodologies, together with in vivo electrophysiology and in vitro approaches on primary cells, were used to afford a comparative and longitudinal analysis of gastrocnemius medialis between fast- and slow-progressing ALS mice. RESULTS: We reported that slow-progressing mice counteracted muscle denervation atrophy by increasing acetylcholine receptor clustering, enhancing evoked currents, and preserving compound muscle action potential. This matched with prompt and sustained myogenesis, likely triggered by an early inflammatory response switching the infiltrated macrophages towards a M2 pro-regenerative phenotype. Conversely, upon denervation, fast-progressing mice failed to promptly activate a compensatory muscle response, exhibiting a rapidly progressive deterioration of muscle force. CONCLUSIONS: Our findings further pinpoint the pivotal role of skeletal muscle in ALS, providing new insights into underestimated disease mechanisms occurring at the periphery and providing useful (diagnostic, prognostic, and mechanistic) information to facilitate the translation of cost-effective therapeutic strategies from the laboratory to the clinic.

13.
J Neurosci ; 31(34): 12330-8, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21865475

RESUMEN

Repeated administration of antipsychotic drugs to normal rats has been shown to induce a state of dopamine neuron inactivation known as depolarization block, which correlates with the ability of the drugs to exhibit antipsychotic efficacy and extrapyramidal side effects in schizophrenia patients. Nonetheless, in normal rats depolarization block requires weeks of antipsychotic drug administration, whereas schizophrenia patients exhibit initial effects soon after initiating antipsychotic drug treatment. We now report that, in a developmental disruption rat model of schizophrenia [methyl-azoxymethanol acetate (20 mg/kg, i.p.) injected into G17 pregnant female rats, with offspring tested as adults], the extant hyperdopaminergic state combines with the excitatory actions of a first- (haloperidol; 0.6 mg/kg, i.p.) and a second- (sertindole; 2.5 mg/kg, i.p.) generation antipsychotic drug to rapidly induce depolarization block in ventral tegmental area dopamine neurons. Acute injection of either antipsychotic drug induced an immediate reduction in the number of spontaneously active dopamine neurons (cells per electrode track; termed population activity). Repeated administration of either antipsychotic drug for 1, 3, 7, 15, and 21 d continued to reduce dopamine neuron population activity. Both acute and repeated effects on population activity were reversed by acute apomorphine injections, which is consistent with the reversal of dopamine neuron depolarization block. Although this action may account for the effects of D2 antagonist drugs on alleviating psychosis and the lack of development of tolerance in humans, the drugs appear to do so by inducing an offsetting deficit rather than attacking the primary pathology present in schizophrenia.


Asunto(s)
Antipsicóticos/farmacología , Dopamina/fisiología , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Área Tegmental Ventral/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Polaridad Celular/efectos de los fármacos , Polaridad Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Acetato de Metilazoximetanol/toxicidad , Inhibición Neural/fisiología , Neuronas/metabolismo , Neurotoxinas/toxicidad , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología
14.
Int J Neuropsychopharmacol ; 15(7): 957-64, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21745437

RESUMEN

Temporal lobe epilepsy (TLE) is defined as the occurrence of spontaneous seizures that involve the limbic system, with the hippocampal formation and associated structures being central to the most prevalent refractory form of adult focal epilepsy. TLE is often associated with psychotic features resembling the hallucinations and delusions that occur with schizophrenia. Given evidence that the ventral hippocampus plays an important role in the maintenance of temporal lobe seizures, we investigated whether an animal model of TLE using intrahippocampal injection of pilocarpine induces alterations in mesolimbic dopamine neuron activity. We found that in 60% of rats in which pilocarpine induced seizure activity, there was a significant increase in the number of dopamine neurons firing per electrode track. Furthermore, this occurred in concert with an increase in amphetamine-stimulated locomotor activity. Both observations are similar to those observed in a rodent developmental model of psychosis. Therefore, as in animal models of schizophrenia, TLE-associated psychosis is probably due to abnormal hippocampal overdrive of dopamine neuron activity.


Asunto(s)
Potenciales de Acción/fisiología , Neuronas Dopaminérgicas/efectos de los fármacos , Epilepsia del Lóbulo Temporal/inducido químicamente , Sistema Límbico/patología , Agonistas Muscarínicos/toxicidad , Pilocarpina/toxicidad , Potenciales de Acción/efectos de los fármacos , Anfetamina , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Dopaminérgicos , Epilepsia del Lóbulo Temporal/patología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Locomoción/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
15.
Life (Basel) ; 12(12)2022 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-36556407

RESUMEN

Recently, the potential use of phytocannabinoids (pCBs) to treat different pathological conditions has attracted great attention in the scientific community. Among the different pCBs, cannabidiol (CBD) has showed interesting biological properties, making it a promising molecule with a high security profile that has been approved for treatment as an add-on therapy in patients afflicted by severe pharmaco-resistant epilepsy, including Dravet syndrome (DS), Lennox-Gastaut syndrome (LGS) and tuberous sclerosis complex (TSC). CBD is pharmacologically considered a "dirty drug", since it has the capacity to bind different targets and to activate several cellular pathways. GABAergic impairment is one of the key processes during the epileptogenesis period able to induce a generalized hyperexcitability of the central nervous system (CNS), leading to epileptic seizures. Here, by using the microtransplantation of human brain membranes approach in Xenopus oocytes and electrophysiological recordings, we confirm the ability of CBD to modulate GABAergic neurotransmission in human cerebral tissues obtained from patients afflicted by different forms of pharmaco-resistant epilepsies, such as DS, TSC, focal cortical dysplasia (FCD) type IIb and temporal lobe epilepsy (TLE). Furthermore, using cDNAs encoding for human GABAA receptor subunits, we found that α1ß2 receptors are still affected by CBD, while classical benzodiazepine lost its efficacy as expected.

16.
Biomolecules ; 12(6)2022 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-35740883

RESUMEN

Recently, the endocannabinoid system has attracted growing attention from the scientific community for its involvement in homeostatic and pathological processes as they pertains to human physiology. Among the constituents of the endocannabinoid system, the molecule palmitoyl ethanolamide has particularly been studied for its ability to reduce several inflammatory processes involving the central nervous system. Here, we reviewed published literature and summarized the main targets of the palmitoyl ethanolamide, along with its unique possible mechanisms for restoring correct functioning of the central nervous system. Moreover, we have highlighted a less-known characteristic of palmitoyl ethanolamide, namely its ability to modulate the function of the neuromuscular junction by binding to acetylcholine receptors in different experimental conditions. Indeed, there are several studies that have highlighted how ultra-micronized palmitoyl ethanolamide is an interesting nutraceutical support for the treatment of pathological neuromuscular conditions, specifically when the normal activity of the acetylcholine receptor is altered. Although further multicentric clinical trials are needed to confirm the efficacy of ultra-micronized palmitoyl ethanolamide in improving symptoms of neuromuscular diseases, all the literature reviewed here strongly supports the ability of this endocannabinoid-like molecule to modulate the acetylcholine receptors thus resulting as a valid support for the treatment of human neuromuscular diseases.


Asunto(s)
Endocannabinoides , Enfermedades Neuromusculares , Receptores Colinérgicos , Endocannabinoides/metabolismo , Humanos , Enfermedades Neuromusculares/tratamiento farmacológico
17.
Front Pharmacol ; 13: 982434, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36052140

RESUMEN

Background: 3,4-diaminopyridine (3,4-DAP) can lead to clinical and electrophysiological improvement in myasthenic syndrome; it may thus represent a valuable therapeutic option for patients intolerant to pyridostigmine. Objective: to assess 3,4-diaminopyridine (3,4-DAP) effects and tolerability in patients with anti-AChR myasthenia gravis. Method: Effects were monitored electrophysiologically by repetitive nerve stimulation (RNS) and by standardized clinical testing (QMG score) before and after a single dose administration of 3,4-DAP 10 mg per os in 15 patients. Patients were divided according to their Myasthenia Gravis Foundation of America (MGFA) class into mild and severe. Results: No significant side effects were found, apart from transient paresthesia. 3,4-DAP had a significant effect on the QMG score (p = 0.0251), on repetitive nerve stimulation (p = 0.0251), and on the forced vital capacity (p = 0.03), thus indicating that it may reduce the level of disability and the decremental muscle response. When the patients were divided according to the MGFA classification, 3,4-DAP showed a positive effect in the severe group, either for the QMG score (p = 0.031) or for the RNS decrement (p = 0.031). No significant difference was observed in any of the outcome measures within the mild group (p > 0.05). A direct effect of 3,4-DAP on nicotinic ACh receptors (nAChRs) was excluded since human nAChRs reconstituted in an expression system, which were not affected by 3,4-DAP application. Conclusion: Our results suggest that 3,4-DAP may be a useful add-on therapy, especially in most severe patients or when immunosuppressive treatment has not yet reached its full effect or when significant side-effects are associated with anticholinesterase.

18.
Brain Sci ; 12(6)2022 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-35741692

RESUMEN

Focal cortical dysplasia (FCD) type II is an epileptogenic malformation of the neocortex, as well as a leading cause of drug-resistant focal epilepsy in children and young adults. The synaptic dysfunctions leading to intractable seizures in this disease appear to have a tight relationship with the immaturity of GABAergic neurotransmission. The likely outcome would include hyperpolarizing responses upon activation of GABAARs. In addition, it is well-established that neuroinflammation plays a relevant role in the pathogenesis of FCD type II. Here, we investigated whether IL-1ß, a prototypical pro-inflammatory cytokine, can influence GABAergic neurotransmission in FCD brain tissues. To this purpose, we carried out electrophysiological recordings on Xenopus oocytes transplanted with human tissues and performed a transcriptomics analysis. We found that IL-1ß decreases the GABA currents amplitude in tissue samples from adult individuals, while it potentiates GABA responses in samples from pediatric cases. Interestingly, these cases of pediatric FCD were characterized by a more depolarized EGABA and an altered transcriptomics profile, that revealed an up-regulation of chloride cotransporter NKCC1 and IL-1ß. Altogether, these results suggest that the neuroinflammatory processes and altered chloride homeostasis can contribute together to increase the brain excitability underlying the occurrence of seizures in these children.

19.
Sci Rep ; 12(1): 17956, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36289354

RESUMEN

Gangliogliomas (GGs) are low-grade brain tumours that cause intractable focal epilepsy in children and adults. In GG, as in epileptogenic focal malformations (i.e., tuberous sclerosis complex, TSC), there is evidence of sustained neuroinflammation with involvement of the pro-inflammatory cytokine IL-1ß. On the other hand, anti-inflammatory mediators are less studied but bear relevance for understanding seizure mechanisms. Therefore, we investigated the effect of the key anti-inflammatory cytokine IL-10 on GABAergic neurotransmission in GG. We assessed the IL-10 dependent signaling by transcriptomic analysis, immunohistochemistry and performed voltage-clamp recordings on Xenopus oocytes microtransplanted with cell membranes from brain specimens, to overcome the limited availability of acute GG slices. We report that IL-10-related mRNAs were up-regulated in GG and slightly in TSC. Moreover, we found IL-10 receptors are expressed by neurons and astroglia. Furthermore, GABA currents were potentiated significantly by IL-10 in GG. This effect was time and dose-dependent and inhibited by blockade of IL-10 signaling. Notably, in the same tissue, IL-1ß reduced GABA current amplitude and prevented the IL-10 effect. These results suggest that in epileptogenic tissue, pro-inflammatory mechanisms of hyperexcitability prevail over key anti-inflammatory pathways enhancing GABAergic inhibition. Hence, boosting the effects of specific anti-inflammatory molecules could resolve inflammation and reduce intractable seizures.


Asunto(s)
Epilepsia Refractaria , Ganglioglioma , Adulto , Niño , Humanos , Ácido gamma-Aminobutírico , Ganglioglioma/complicaciones , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Receptores de GABA-A/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Interleucina-10/metabolismo
20.
Biomedicines ; 10(5)2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35625812

RESUMEN

Mutations in SCN1A gene, encoding the voltage-gated sodium channel (VGSC) NaV1.1, are widely recognized as a leading cause of genetic febrile seizures (FS), due to the decrease in the Na+ current density, mainly affecting the inhibitory neuronal transmission. Here, we generated induced pluripotent stem cells (iPSCs)-derived neurons (idNs) from a patient belonging to a genetically well-characterized Italian family, carrying the c.434T > C mutation in SCN1A gene (hereafter SCN1AM145T). A side-by-side comparison of diseased and healthy idNs revealed an overall maturation delay of SCN1AM145T cells. Membranes isolated from both diseased and control idNs were injected into Xenopus oocytes and both GABA and AMPA currents were successfully recorded. Patch-clamp measurements on idNs revealed depolarized action potential for SCN1AM145T, suggesting a reduced excitability. Expression analyses of VGSCs and chloride co-transporters NKCC1 and KCC2 showed a cellular "dysmaturity" of mutated idNs, strengthened by the high expression of SCN3A, a more fetal-like VGSC isoform, and a high NKCC1/KCC2 ratio, in mutated cells. Overall, we provide strong evidence for an intrinsic cellular immaturity, underscoring the role of mutant NaV1.1 in the development of FS. Furthermore, our data are strengthening previous findings obtained using transfected cells and recordings on human slices, demonstrating that diseased idNs represent a powerful tool for personalized therapy and ex vivo drug screening for human epileptic disorders.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA