Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 585(7825): 420-425, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32879486

RESUMEN

The opsin family of G-protein-coupled receptors are used as light detectors in animals. Opsin 5 (also known as neuropsin or OPN5) is a highly conserved opsin that is sensitive to visible violet light1,2. In mice, OPN5 is a known photoreceptor in the retina3 and skin4 but is also expressed in the hypothalamic preoptic area (POA)5. Here we describe a light-sensing pathway in which POA neurons that express Opn5 regulate thermogenesis in brown adipose tissue (BAT). We show that Opn5 is expressed in glutamatergic warm-sensing POA neurons that receive synaptic input from several thermoregulatory nuclei. We further show that Opn5 POA neurons project to BAT and decrease its activity under chemogenetic stimulation. Opn5-null mice show overactive BAT, increased body temperature, and exaggerated thermogenesis when cold-challenged. Moreover, violet photostimulation during cold exposure acutely suppresses BAT temperature in wild-type mice but not in Opn5-null mice. Direct measurements of intracellular cAMP ex vivo show that Opn5 POA neurons increase cAMP when stimulated with violet light. This analysis thus identifies a violet light-sensitive deep brain photoreceptor that normally suppresses BAT thermogenesis.


Asunto(s)
Color , Luz , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Neuronas/efectos de la radiación , Opsinas/metabolismo , Área Preóptica/citología , Termogénesis/efectos de la radiación , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/efectos de la radiación , Animales , Temperatura Corporal , Frío , AMP Cíclico/metabolismo , Femenino , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Opsinas/deficiencia , Opsinas/genética , Termogénesis/genética
2.
Neurobiol Dis ; 144: 105026, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32712265

RESUMEN

Epilepsy affects all ages, races, genders, and socioeconomic groups. In about one third of patients, epilepsy is uncontrolled with current medications, leaving a vast need for improved therapies. The causes of epilepsy are diverse and not always known but one gene mutated in a small subpopulation of patients is phosphatase and tensin homolog (PTEN). Moreover, focal cortical dysplasia, which constitutes a large fraction of refractory epilepsies, has been associated with signaling defects downstream of PTEN. So far, most preclinical attempts to reverse PTEN deficiency-associated neurological deficits have focused on mTOR, a signaling hub several steps downstream of PTEN. Phosphoinositide 3-kinases (PI3Ks), by contrast, are the direct enzymatic counteractors of PTEN, and thus may be alternative treatment targets. PI3K activity is mediated by four different PI3K catalytic isoforms. Studies in cancer, where PTEN is commonly mutated, have demonstrated that inhibition of only one isoform, p110ß, reduces progression of PTEN-deficient tumors. Importantly, inhibition of a single PI3K isoform leaves critical functions of general PI3K signaling throughout the body intact. Here, we show that this disease mechanism-targeted strategy borrowed from cancer research rescues or ameliorates neuronal phenotypes in male and female mice with neuron-specific PTEN deficiency. These phenotypes include cell signaling defects, protein synthesis aberrations, seizures, and cortical dysplasia. Of note, p110ß is also dysregulated and a promising treatment target in the intellectual disability Fragile X syndrome, pointing towards a shared biological mechanism that is therapeutically targetable in neurodevelopmental disorders of different etiologies. Overall, this work advocates for further assessment of p110ß inhibition not only in PTEN deficiency-associated neurodevelopmental diseases but also other brain disorders characterized by defects in the PI3K/mTOR pathway.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Epilepsia/fisiopatología , Neuronas/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Epilepsia/genética , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Megalencefalia/fisiopatología , Ratones , Neuronas/metabolismo , Fosfohidrolasa PTEN/genética , Quinazolinas/farmacología , Convulsiones/fisiopatología , Tiazoles/farmacología
3.
Neurobiol Dis ; 130: 104508, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31212067

RESUMEN

Epilepsy is often associated with altered expression or function of ion channels. One example of such a channelopathy is the reduction of A-type potassium currents in the hippocampal CA1 region. The underlying mechanisms of reduced A-type channel function in epilepsy are unclear. Here, we show that inhibiting a single microRNA, miR-324-5p, which targets the pore-forming A-type potassium channel subunit Kv4.2, selectively increased A-type potassium currents in hippocampal CA1 pyramidal neurons in mice. Resting membrane potential, input resistance and other potassium currents were not altered. In a mouse model of acquired chronic epilepsy, inhibition of miR-324-5p reduced the frequency of spontaneous seizures and interictal epileptiform spikes supporting the physiological relevance of miR-324-5p-mediated control of A-type currents in regulating neuronal excitability. Mechanistic analyses demonstrated that microRNA-induced silencing of Kv4.2 mRNA is increased in epileptic mice leading to reduced Kv4.2 protein levels, which is mitigated by miR-324-5p inhibition. By contrast, other targets of miR-324-5p were unchanged. These results suggest a selective miR-324-5p-dependent mechanism in epilepsy regulating potassium channel function, hyperexcitability and seizures.


Asunto(s)
Epilepsia/fisiopatología , Hipocampo/fisiopatología , MicroARNs/metabolismo , Convulsiones/fisiopatología , Canales de Potasio Shal/metabolismo , Regulación hacia Arriba , Animales , Modelos Animales de Enfermedad , Epilepsia/metabolismo , Hipocampo/metabolismo , Ratones , MicroARNs/genética , Convulsiones/metabolismo , Canales de Potasio Shal/genética
4.
Dev Dyn ; 247(1): 94-110, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28850760

RESUMEN

MicroRNAs are master regulators of gene expression. Single microRNAs influence multiple proteins within diverse molecular pathways and networks. Therefore, changes in levels or activity of microRNAs can have profound effects on cellular function. This makes dysregulated microRNA-induced silencing an attractive potential disease mechanism in complex disorders like epilepsy, where numerous cellular pathways and processes are affected simultaneously. Indeed, several years of research in rodent models have provided strong evidence that acute or recurrent seizures change microRNA expression and function. Moreover, altered microRNA expression has been observed in brain and blood from patients with various epilepsy disorders, such as tuberous sclerosis. MicroRNAs can be easily manipulated using sense or antisense oligonucleotides, opening up opportunities for therapeutic intervention. Here, we summarize studies using these techniques to identify microRNAs that modulate seizure susceptibility, describe protein targets mediating some of these effects, and discuss cellular pathways, for example neuroinflammation, that are controlled by epilepsy-associated microRNAs. We critically assess current gaps in knowledge regarding target- and cell-specificity of microRNAs that have to be addressed before clinical application as therapeutic targets or biomarkers. The recent progress in understanding microRNA function in epilepsy has generated strong momentum to encourage in-depth mechanistic studies to develop microRNA-targeted therapies. Developmental Dynamics 247:94-110, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Encéfalo/metabolismo , Epilepsia/terapia , Silenciador del Gen , MicroARNs/genética , Oligonucleótidos Antisentido/uso terapéutico , Animales , Epilepsia/genética , Humanos
5.
Brain Behav Immun ; 70: 36-47, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29545118

RESUMEN

Epidemiological evidence suggests that people with bipolar disorder prescribed lithium exhibit a lower risk of Alzheimer's disease (AD) relative to those prescribed other mood-stabilizing medicines. Lithium chloride (LiCl) reduces brain ß-amyloid (Aß) levels, and the brain clearance of Aß is reduced in AD. Therefore, the purpose of this study was to assess whether the cognitive benefits of LiCl are associated with enhanced brain clearance of exogenously-administered Aß. The brain clearance of intracerebroventricularly (icv) administered 125I-Aß42 was assessed in male Swiss outbred mice administered daily oral NaCl or LiCl (300 mg/kg for 21 days). LiCl exhibited a 31% increase in the brain clearance of 125I-Aß42 over 10 min, which was associated with a 1.6-fold increase in brain microvascular expression of the blood-brain barrier efflux transporter low density lipoprotein receptor-related protein 1 (LRP1) and increased cerebrospinal fluid (CSF) bulk-flow. 8-month-old female wild type (WT) and APP/PS1 mice were also administered daily NaCl or LiCl for 21 days, which was followed by cognitive assessment by novel object recognition and water maze, and measurement of soluble Aß42, plaque-associated Aß42, and brain efflux of 125I-Aß42. LiCl treatment restored the long-term spatial memory deficit observed in APP/PS1 mice as assessed by the water maze (back to similar levels of escape latency as WT mice), but the short-term memory deficit remained unaffected by LiCl treatment. While LiCl did not affect plaque-associated Aß42, soluble Aß42 levels were reduced by 49.9% in APP/PS1 mice receiving LiCl. The brain clearance of 125I-Aß42 decreased by 27.8% in APP/PS1 mice, relative to WT mice, however, LiCl treatment restored brain 125I-Aß42 clearance in APP/PS1 mice to a rate similar to that observed in WT mice. These findings suggest that the cognitive benefits and brain Aß42 lowering effects of LiCl are associated with enhanced brain clearance of Aß42, possibly via brain microvascular LRP1 upregulation and increased CSF bulk-flow, identifying a novel mechanism of protection by LiCl for the treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Cognición/efectos de los fármacos , Cloruro de Litio/uso terapéutico , Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo , Modelos Animales de Enfermedad , Cloruro de Litio/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Transgénicos , Placa Amiloide , Presenilina-1 , Receptores de LDL/efectos de los fármacos , Receptores de LDL/fisiología , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/fisiología
6.
Curr Neurol Neurosci Rep ; 18(9): 60, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30046905

RESUMEN

PURPOSE OF REVIEW: The goal of this focused review is to describe recent studies supporting a critical role of microRNAs in the regulation of ion channels and discuss the resulting implications for the modulation of neuronal excitability in epilepsy. RECENT FINDINGS: MicroRNA-induced silencing of ion channels has been shown in several different studies in recent years, and some of these reports suggest a prominent role in epilepsy. The ion channels regulated by microRNAs include ligand- and voltage-gated channels and are not only limited to the central nervous system but have also been found in the peripheral nervous system. Ion channel-targeting microRNAs can regulate the intrinsic excitability of neurons, and thus influence entire networks in the brain. Their dysregulation in epilepsy may contribute to the disease phenotype. More research is needed to better understand the molecular mechanisms of how microRNAs regulate ion channels to control neuronal excitability, and how these processes are altered in epilepsy.


Asunto(s)
Epilepsia/genética , Epilepsia/terapia , Terapia Genética/métodos , Activación del Canal Iónico/fisiología , MicroARNs/fisiología , Animales , Terapia Genética/tendencias , Humanos , Canales Iónicos/fisiología , MicroARNs/uso terapéutico , Neuronas/fisiología
7.
Cureus ; 15(10): e47777, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38022199

RESUMEN

Introduction The phenomenon of coronavirus disease 2019 (COVID-19)-related candidiasis is gaining increased attention and acknowledgment as an integral component of the severe consequences of COVID-19. The aim of the present study was to assess the association between Candida albicans and COVID-19 in complete denture wearers. Materials and methods An observational study was conducted on 45 complete denture wearers, who were divided into three groups as follows: Group 1, 15 subjects with mild to moderate COVID-19 infection; Group 2, 15 subjects with severe COVID-19 infection; and Group 3, 15 subjects without COVID-19 infection. Mean colony forming units (CFU) were observed on agar plates containing Sabouraud dextrose in the salivary samples of the participants. Analysis of variance, followed by post-hoc analysis by Tukey's test, was used to compare CFU between the groups. Pearson's correlation coefficient was used to study the correlation between variables. Results The highest average colony-forming units of Candida albicans were observed in Group 2, followed by Group 1, compared to the control group, and a significant (p<0.001) difference was found. A weak positive correlation was found between the age of the patients and the duration of denture usage, as well as between age and the counts of Candida albicans in Groups 1 and 3. This correlation was more pronounced in Group 3. A strong positive correlation was observed in all groups between the Candida albicans count and the duration of denture usage by the patients. Conclusion The association between Candida albicans and denture wear was compounded by the presence of COVID-19. Consequently, the timely identification of Candida albicans infection in patients with COVID-19 is important to establish more efficacious approaches for antifungal treatment and prophylactic interventions.

8.
Res Sq ; 2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-37609225

RESUMEN

Dendritic spines are small, dynamic protrusions along the dendrite that comprise more than 90% of excitatory connections in the brain, making them essential sites for neuronal communication. These synaptic sites change throughout the process of development, reducing in density and shifting morphology as synapses are refined. One important class of dendritic spine regulators is microRNA (miRNA), small noncoding RNAs that post-transcriptionally regulate gene expression. Several studies suggest that miRNA-324-5p regulates dendritic spine formation. In addition, we have previously shown that miR-324-5p plays a role in seizure and long-term potentiation, both of which involve dendritic spine changes. In this study, we aimed to characterize the role of miRNA-324-5p in developmental spine regulation by assessing the effect of Mir324 knockout (KO) on dendritic spine density and expression of a subset of dendritic proteins at select developmental time points. We show that miR-324-5p expression is developmentally regulated and peaks at four weeks of age. We demonstrate that loss of miR-324-5p expression leads to differential changes in both target protein expression and spine density at different time points during development, disrupting the pattern of spine density changes and leading to a premature loss of dendritic spines in KO mice, which is compensated later. Our findings indicate that miR-324-5p plays a role in synaptic refinement across development. Additionally, our data illustrate the importance of context in the study of miRNA, as regulation by and/or of miRNA can vary dramatically across development and in disease.

9.
Sci Rep ; 13(1): 21919, 2023 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-38082035

RESUMEN

MicroRNAs are an emerging class of synaptic regulators. These small noncoding RNAs post-transcriptionally regulate gene expression, thereby altering neuronal pathways and shaping cell-to-cell communication. Their ability to rapidly alter gene expression and target multiple pathways makes them interesting candidates in the study of synaptic plasticity. Here, we demonstrate that the proconvulsive microRNA miR-324-5p regulates excitatory synapse structure and function in the hippocampus of mice. Both Mir324 knockout (KO) and miR-324-5p antagomir treatment significantly reduce dendritic spine density in the hippocampal CA1 subregion, and Mir324 KO, but not miR-324-5p antagomir treatment, shift dendritic spine morphology, reducing the proportion of thin, "unstable" spines. Western blot and quantitative Real-Time PCR revealed changes in protein and mRNA levels for potassium channels, cytoskeletal components, and synaptic markers, including MAP2 and Kv4.2, which are important for long-term potentiation (LTP). In line with these findings, slice electrophysiology revealed that LTP is severely impaired in Mir324 KO mice, while neurotransmitter release probability remains unchanged. Overall, this study demonstrates that miR-324-5p regulates dendritic spine density, morphology, and plasticity in the hippocampus, potentially via multiple cytoskeletal and synaptic modulators.


Asunto(s)
Potenciación a Largo Plazo , MicroARNs , Ratones , Animales , Potenciación a Largo Plazo/fisiología , Espinas Dendríticas/metabolismo , Antagomirs/metabolismo , Hipocampo/metabolismo , Plasticidad Neuronal/genética , Sinapsis/metabolismo , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo
10.
J Mol Neurosci ; 73(9-10): 818-830, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37773316

RESUMEN

Dendritic spines are small, dynamic protrusions along the dendrite that comprise more than 90% of excitatory connections in the brain, making them essential sites for neuronal communication. These synaptic sites change throughout the process of development, reducing in density and shifting morphology as synapses are refined. One important class of dendritic spine regulators is microRNA (miRNA), small-noncoding RNAs that post-transcriptionally regulate gene expression. Several studies suggest that miRNA-324-5p regulates dendritic spine formation. In addition, we have previously shown that miR-324-5p plays a role in seizure and long-term potentiation, both of which involve dendritic spine changes. In this study, we aimed to characterize the role of miRNA-324-5p in developmental spine regulation by assessing the effect of Mir324 knockout (KO) on dendritic spine density and expression of a subset of dendritic proteins at select developmental time points. We show that miR-324-5p expression is developmentally regulated and peaks at 4 weeks of age. We demonstrate that loss of miR-324-5p expression leads to differential changes in both target protein expression and spine density at different time points during development, disrupting the pattern of spine density changes and leading to a premature loss of dendritic spines in KO mice, which is compensated later. Our findings indicate that miR-324-5p plays a role in synaptic refinement across development. Additionally, our data illustrate the importance of context in the study of miRNA, as regulation by and/or of miRNA can vary dramatically across development and in disease.


Asunto(s)
Espinas Dendríticas , MicroARNs , Animales , Ratones , Espinas Dendríticas/metabolismo , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo
11.
eNeuro ; 10(7)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37433683

RESUMEN

About one-third of individuals living with epilepsy have treatment-resistant seizures. Alternative therapeutic strategies are thus urgently needed. One potential novel treatment target is miRNA-induced silencing, which is differentially regulated in epilepsy. Inhibitors (antagomirs) of specific microRNAs (miRNAs) have shown therapeutic promise in preclinical epilepsy studies; however, these studies were mainly conducted in male rodent models, and research into miRNA regulation in females and by female hormones in epilepsy is scarce. This is problematic because female sex and the menstrual cycle can affect the disease course of epilepsy and may, therefore, also alter the efficacy of potential miRNA-targeted treatments. Here, we used the proconvulsant miRNA miR-324-5p and its target, the potassium channel Kv4.2, as an example to test how miRNA-induced silencing and the efficacy of antagomirs in epilepsy are altered in female mice. We showed that Kv4.2 protein is reduced after seizures in female mice similar to male mice; however, in contrast to male mice, miRNA-induced silencing of Kv4.2 is unchanged, and miR-324-5p activity, as measured by the association with the RNA-induced silencing complex, is reduced in females after seizure. Moreover, an miR-324-5p antagomir does not consistently reduce seizure frequency or increase Kv4.2 in female mice. As a possible underlying mechanism, we found that miR-324-5p activity and the silencing of Kv4.2 in the brain were differentially correlated with plasma levels of 17ß-estradiol and progesterone. Our results suggest that hormonal fluctuations in sexually mature female mice influence miRNA-induced silencing and could alter the efficacy of potential future miRNA-based treatments for epilepsy in females.


Asunto(s)
Epilepsia , MicroARNs , Ratones , Masculino , Femenino , Animales , MicroARNs/genética , Antagomirs/farmacología , Progesterona/metabolismo , Estradiol/metabolismo , Hipocampo/metabolismo , Modelos Animales de Enfermedad , Convulsiones/inducido químicamente , Epilepsia/metabolismo
12.
Front Neurol ; 14: 1280606, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38033777

RESUMEN

Background: Acquired epilepsies are caused by an initial brain insult that is followed by epileptogenesis and finally the development of spontaneous recurrent seizures. The mechanisms underlying epileptogenesis are not fully understood. MicroRNAs regulate mRNA translation and stability and are frequently implicated in epilepsy. For example, antagonism of a specific microRNA, miR-324-5p, before brain insult and in a model of chronic epilepsy decreases seizure susceptibility and frequency, respectively. Here, we tested whether antagonism of miR-324-5p during epileptogenesis inhibits the development of epilepsy. Methods: We used the intrahippocampal kainic acid (IHpKa) model to initiate epileptogenesis in male wild type C57BL/6 J mice aged 6-8 weeks. Twenty-four hours after IHpKa, we administered a miR-324-5p or scrambled control antagomir intracerebroventricularly and implanted cortical surface electrodes for EEG monitoring. EEG data was collected for 28 days and analyzed for seizure frequency and duration, interictal spike activity, and EEG power. Brains were collected for histological analysis. Results: Histological analysis of brain tissue showed that IHpKa caused characteristic hippocampal damage in most mice regardless of treatment. Antagomir treatment did not affect latency to, frequency, or duration of spontaneous recurrent seizures or interictal spike activity but did alter the temporal development of frequency band-specific EEG power. Conclusion: These results suggest that miR-324-5p inhibition during epileptogenesis induced by status epilepticus does not convey anti-epileptogenic effects despite having subtle effects on EEG frequency bands. Our results highlight the importance of timing of intervention across epilepsy development and suggest that miR-324-5p may act primarily as a proconvulsant rather than a pro-epileptogenic regulator.

13.
Elife ; 122023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37862092

RESUMEN

The assembly of the mammalian brain is orchestrated by temporally coordinated waves of gene expression. Post-transcriptional regulation by microRNAs (miRNAs) is a key aspect of this program. Indeed, deletion of neuron-enriched miRNAs induces strong developmental phenotypes, and miRNA levels are altered in patients with neurodevelopmental disorders. However, the mechanisms used by miRNAs to instruct brain development remain largely unexplored. Here, we identified miR-218 as a critical regulator of hippocampal assembly. MiR-218 is highly expressed in the hippocampus and enriched in both excitatory principal neurons (PNs) and GABAergic inhibitory interneurons (INs). Early life inhibition of miR-218 results in an adult brain with a predisposition to seizures. Changes in gene expression in the absence of miR-218 suggest that network assembly is impaired. Indeed, we find that miR-218 inhibition results in the disruption of early depolarizing GABAergic signaling, structural defects in dendritic spines, and altered intrinsic membrane excitability. Conditional knockout of Mir218-2 in INs, but not PNs, is sufficient to recapitulate long-term instability. Finally, de-repressing Kif21b and Syt13, two miR-218 targets, phenocopies the effects on early synchronous network activity induced by miR-218 inhibition. Taken together, the data suggest that miR-218 orchestrates formative events in PNs and INs to produce stable networks.


Asunto(s)
MicroARNs , Animales , Adulto , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/metabolismo , Hipocampo/metabolismo , Interneuronas/metabolismo , Encéfalo/metabolismo , Mamíferos/genética
14.
Prog Neurobiol ; 200: 101974, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33309800

RESUMEN

Mutations in genes regulating mTOR pathway signaling are now recognized as a significant cause of epilepsy. Interestingly, these mTORopathies are often caused by somatic mutations, affecting variable numbers of neurons. To better understand how this variability affects disease phenotype, we developed a mouse model in which the mTOR pathway inhibitor Pten can be deleted from 0 to 40 % of hippocampal granule cells. In vivo, low numbers of knockout cells caused focal seizures, while higher numbers led to generalized seizures. Generalized seizures coincided with the loss of local circuit interneurons. In hippocampal slices, low knockout cell loads produced abrupt reductions in population spike threshold, while spontaneous excitatory postsynaptic currents and circuit level recurrent activity increased gradually with rising knockout cell load. Findings demonstrate that knockout cells load is a critical variable regulating disease phenotype, progressing from subclinical circuit abnormalities to electrobehavioral seizures with secondary involvement of downstream neuronal populations.


Asunto(s)
Epilepsia , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones , Neuronas/metabolismo , Convulsiones , Serina-Treonina Quinasas TOR/metabolismo
15.
Front Psychiatry ; 12: 678090, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093287

RESUMEN

Fragile X syndrome (FXS) is the most common cause of inherited intellectual disability. FXS is caused by functional loss of the Fragile X Protein (FXP), also known as Fragile X Mental Retardation Protein (FMRP). In humans and animal models, loss of FXP leads to sensory hypersensitivity, increased susceptibility to seizures and cortical hyperactivity. Several components of the GABAergic system, the major inhibitory system in the brain, are dysregulated in FXS, and thus modulation of GABAergic transmission was suggested and tested as a treatment strategy. However, so far, clinical trials using broad spectrum GABAA or GABAB receptor-specific agonists have not yielded broad improvement of FXS phenotypes in humans. Here, we tested a more selective strategy in Fmr1 knockout (KO) mice using the experimental drug BAER-101, which is a selective GABAA α2/α3 agonist. Our results suggest that BAER-101 reduces hyperexcitability of cortical circuits, partially corrects increased frequency-specific baseline cortical EEG power, reduces susceptibility to audiogenic seizures and improves novel object memory. Other Fmr1 KO-specific phenotypes were not improved by the drug, such as increased hippocampal dendritic spine density, open field activity and marble burying. Overall, this work shows that BAER-101 improves select phenotypes in Fmr1 KO mice and encourages further studies into the efficacy of GABAA-receptor subunit-selective agonists for the treatment of FXS.

16.
Exp Neurol ; 334: 113437, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32822706

RESUMEN

The voltage-gated potassium channel Kv4.2 is a critical regulator of dendritic excitability in the hippocampus and is crucial for dendritic signal integration. Kv4.2 mRNA and protein expression as well as function are reduced in several genetic and pharmacologically induced rodent models of epilepsy and autism. It is not known, however, whether reduced Kv4.2 is just an epiphenomenon or a disease-contributing cause of neuronal hyperexcitability and behavioral impairments in these neurological disorders. To address this question, we used male and female mice heterozygous for a Kv.2 deletion and adult-onset manipulation of hippocampal Kv4.2 expression in male mice to assess the role of Kv4.2 in regulating neuronal network excitability, morphology and anxiety-related behaviors. We observed a reduction in dendritic spine density and reduced proportions of thin and stubby spines but no changes in anxiety, overall activity, or retention of conditioned freezing memory in Kv4.2 heterozygous mice compared with wildtype littermates. Using EEG analyses, we showed elevated theta power and increased spike frequency in Kv4.2 heterozygous mice under basal conditions. In addition, the latency to onset of kainic acid-induced seizures was significantly shortened in Kv4.2 heterozygous mice compared with wildtype littermates, which was accompanied by a significant increase in theta power. By contrast, overexpressing Kv4.2 in wildtype mice through intrahippocampal injection of Kv4.2-expressing lentivirus delayed seizure onset and reduced EEG power. These results suggest that Kv4.2 is an important regulator of neuronal network excitability and dendritic spine morphology, but not anxiety-related behaviors. In the future, manipulation of Kv4.2 expression could be used to alter seizure susceptibility in epilepsy.


Asunto(s)
Espinas Dendríticas/metabolismo , Electroencefalografía/métodos , Hipocampo/metabolismo , Convulsiones/metabolismo , Canales de Potasio Shal/biosíntesis , Animales , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Hipocampo/citología , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Convulsiones/genética , Convulsiones/fisiopatología , Canales de Potasio Shal/genética
17.
Cell Rep ; 32(9): 108092, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32877674

RESUMEN

Lactate is used as an energy source by producer cells or shuttled to neighboring cells and tissues. Both glucose and lactate fulfill the bioenergetic demand of neurons, the latter imported from astrocytes. The contribution of astrocytic lactate to neuronal bioenergetics and the mechanisms of astrocytic lactate production are incompletely understood. Through in vivo1H magnetic resonance spectroscopy, 13C glucose mass spectroscopy, and electroencephalographic and molecular studies, here we show that the energy sensor AMP activated protein kinase (AMPK) regulates neuronal survival in a non-cell-autonomous manner. Ampk-null mice are deficient in brain lactate and are seizure prone. Ampk deletion in astroglia, but not neurons, causes neuronal loss in both mammalian and fly brains. Mechanistically, astrocytic AMPK phosphorylated and destabilized thioredoxin-interacting protein (TXNIP), enabling expression and surface translocation of the glucose transporter GLUT1, glucose uptake, and lactate production. Ampk loss in astrocytes causes TXNIP hyperstability, GLUT1 misregulation, inadequate glucose metabolism, and neuronal loss.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Astrocitos/metabolismo , Ácido Láctico/metabolismo , Neuronas/metabolismo , Animales , Muerte Celular , Humanos , Ratones
18.
Neuropsychopharmacology ; 44(2): 324-333, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30061744

RESUMEN

Defects in the phosphoinositide 3-kinase (PI3K) pathway are shared characteristics in several brain disorders, including the inherited intellectual disability and autism spectrum disorder, fragile X syndrome (FXS). PI3K signaling therefore could serve as a therapeutic target for FXS and other brain disorders. However, broad inhibition of such a central signal transduction pathway involved in essential cellular functions may produce deleterious side effects. Pharmacological strategies that selectively correct the overactive components of the PI3K pathway while leaving other parts of the pathway intact may overcome these challenges. Here, we provide the first evidence that disease mechanism-based PI3K isoform-specific inhibition may be a viable treatment option for FXS. FXS is caused by loss of the fragile X mental retardation protein (FMRP), which translationally represses specific messenger RNAs, including the PI3K catalytic isoform p110ß. FMRP deficiency increases p110ß protein levels and activity in FXS mouse models and in cells from subjects with FXS. Here, we show that a novel, brain-permeable p110ß-specific inhibitor, GSK2702926A, ameliorates FXS-associated phenotypes on molecular, cellular, behavioral, and cognitive levels in two different FMRP-deficient mouse models. Rescued phenotypes included increased PI3K downstream signaling, protein synthesis rates, and dendritic spine density, as well as impaired social interaction and higher-order cognition. Several p110ß-selective inhibitors, for example, a molecule from the same chemotype as GSK2702926A, are currently being evaluated in clinical trials to treat cancer. Our results suggest that repurposing p110ß inhibitors to treat cognitive and behavioral defects may be a promising disease-modifying strategy for FXS and other brain disorders.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Espinas Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Ratones , Actividad Motora/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA