Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37720016

RESUMEN

Neurodevelopmental disorders are frequently linked to mutations in synaptic organizing molecules. MAM domain containing glycosylphosphatidylinositol anchor 1 and 2 (MDGA1 and MDGA2) are a family of synaptic organizers suggested to play an unusual role as synaptic repressors, but studies offer conflicting evidence for their localization. Using epitope-tagged MDGA1 and MDGA2 knock-in mice, we found that native MDGAs are expressed throughout the brain, peaking early in postnatal development. Surprisingly, endogenous MDGA1 was enriched at excitatory, but not inhibitory, synapses. Both shRNA knockdown and CRISPR/Cas9 knockout of MDGA1 resulted in cell-autonomous, specific impairment of AMPA receptor-mediated synaptic transmission, without affecting GABAergic transmission. Conversely, MDGA2 knockdown/knockout selectively depressed NMDA receptor-mediated transmission but enhanced inhibitory transmission. Our results establish that MDGA2 acts as a synaptic repressor, but only at inhibitory synapses, whereas both MDGAs are required for excitatory transmission. This nonoverlapping division of labor between two highly conserved synaptic proteins is unprecedented.

2.
Cell Rep ; 41(2): 111483, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36223737

RESUMEN

The critical role of AMPA receptor (AMPAR) trafficking in long-term potentiation (LTP) of excitatory synaptic transmission is now well established, but the underlying molecular mechanism is still uncertain. Recent research suggests that PSD-95 captures AMPARs via an interaction with the AMPAR auxiliary subunits-transmembrane AMPAR regulatory proteins (TARPs). To determine if such interaction is a core minimal component of the AMPAR trafficking and LTP mechanism, we engineered artificial binding partners, which individually were biochemically and functionally dead but which, when expressed together, rescue binding and both basal synaptic transmission and LTP. These findings establish the TARP/PSD-95 complex as an essential interaction underlying AMPAR trafficking and LTP.


Asunto(s)
Potenciación a Largo Plazo , Receptores AMPA , Homólogo 4 de la Proteína Discs Large/metabolismo , Potenciación a Largo Plazo/fisiología , Proteínas Nucleares/metabolismo , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
3.
Elife ; 102021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34908526

RESUMEN

Long-term potentiation (LTP) is arguably the most compelling cellular model for learning and memory. While the mechanisms underlying the induction of LTP ('learning') are well understood, the maintenance of LTP ('memory') has remained contentious over the last 20 years. Here, we find that Ca2+-calmodulin-dependent kinase II (CaMKII) contributes to synaptic transmission and is required LTP maintenance. Acute inhibition of CaMKII erases LTP and transient inhibition of CaMKII enhances subsequent LTP. These findings strongly support the role of CaMKII as a molecular storage device.


How the brain stores information is a question that has fascinated neuroscientists for well over a century. Two general ideas have emerged. The first is that groups of neurons hold information by staying active. The second is that they hold information by strengthening their connections to one another, making it easier for them to work together in the future. Scientists call this second idea 'long-term potentiation'. One of the molecules involved in long-term potentiation is a protein called calcium-calmodulin-dependent kinase II, or CaMKII for short. Blocking CaMKII, or deleting its gene, stops the connections between neurons from becoming stronger. This suggests neurons need CaMKII to learn, but it remains unclear whether neurons also use CaMKII to maintain neuronal memories after they have been created. If CaMKII does play a role in maintaining memories, blocking it after learning should reverse the learning process, but so far, experiments have not been able to show this. Tao et al. revisited these experiments to find out more. They examined slices of brain tissue from mice that had been treated with fast-acting CaMKII inhibitors. It took tens of minutes, but the inhibitors were able to reverse long-term potentiation, both for newly acquired neuronal memories and for older memories that had formed when the mice were alive. The choice of CaMKII inhibitor and the time lag could explain why scientists have not observed the effect before. Understanding long-term potentiation is a fundamental part of understanding learning and memory. It could also reveal more about the opposite phenomenon: long-term depression. This is a type of learning where the connections between neurons become weaker. Long-term depression also takes tens of minutes to occur, suggesting that future research into CaMKII might shed light on how it works.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Potenciación a Largo Plazo , Transmisión Sináptica , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ratones
4.
Neuropharmacology ; 197: 108710, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34271016

RESUMEN

AMPA receptors (AMPARs) are fundamental elements in excitatory synaptic transmission and synaptic plasticity in the CNS. Long term potentiation (LTP), a form of synaptic plasticity which contributes to learning and memory formation, relies on the accumulation of AMPARs at the postsynapse. This phenomenon requires the coordinated recruitment of different elements in the AMPAR complex. Based on recent research reviewed herein, we propose an updated AMPAR trafficking and LTP model which incorporates both extracellular as well as intracellular mechanisms. This article is part of the special Issue on 'Glutamate Receptors - AMPA receptors'.


Asunto(s)
Potenciación a Largo Plazo/fisiología , Receptores AMPA/fisiología , Animales , Humanos , Potenciación a Largo Plazo/genética , Receptores AMPA/genética , Receptores AMPA/metabolismo
5.
Development ; 147(24)2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33168583

RESUMEN

The endocannabinoid (eCB) system, via the cannabinoid CB1 receptor, regulates neurodevelopment by controlling neural progenitor proliferation and neurogenesis. CB1 receptor signalling in vivo drives corticofugal deep layer projection neuron development through the regulation of BCL11B and SATB2 transcription factors. Here, we investigated the role of eCB signalling in mouse pluripotent embryonic stem cell-derived neuronal differentiation. Characterization of the eCB system revealed increased expression of eCB-metabolizing enzymes, eCB ligands and CB1 receptors during neuronal differentiation. CB1 receptor knockdown inhibited neuronal differentiation of deep layer neurons and increased upper layer neuron generation, and this phenotype was rescued by CB1 re-expression. Pharmacological regulation with CB1 receptor agonists or elevation of eCB tone with a monoacylglycerol lipase inhibitor promoted neuronal differentiation of deep layer neurons at the expense of upper layer neurons. Patch-clamp analyses revealed that enhancing cannabinoid signalling facilitated neuronal differentiation and functionality. Noteworthy, incubation with CB1 receptor agonists during human iPSC-derived cerebral organoid formation also promoted the expansion of BCL11B+ neurons. These findings unveil a cell-autonomous role of eCB signalling that, via the CB1 receptor, promotes mouse and human deep layer cortical neuron development.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Neuronas/metabolismo , Receptor Cannabinoide CB1/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Animales , Proliferación Celular/efectos de los fármacos , Cerebelo/crecimiento & desarrollo , Desarrollo Embrionario/genética , Endocannabinoides/agonistas , Endocannabinoides/genética , Endocannabinoides/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Organoides/crecimiento & desarrollo , Transducción de Señal/genética
6.
Elife ; 92020 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-32831170

RESUMEN

We tested the proposal that the C-terminal domain (CTD) of the AMPAR subunit GluA1 is required for LTP. We found that a knock-in mouse lacking the CTD of GluA1 expresses normal LTP and spatial memory, assayed by the Morris water maze. Our results support a model in which LTP generates synaptic slots, which capture passively diffusing AMPARs.


Asunto(s)
Potenciación a Largo Plazo , Receptores AMPA , Animales , Línea Celular , Femenino , Técnicas de Sustitución del Gen , Hipocampo/metabolismo , Humanos , Potenciación a Largo Plazo/genética , Potenciación a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Dominios Proteicos/genética , Receptores AMPA/química , Receptores AMPA/genética , Receptores AMPA/metabolismo , Memoria Espacial/fisiología
7.
Neuron ; 104(3): 529-543.e6, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492534

RESUMEN

Transmembrane AMPA receptor (AMPAR) regulatory proteins (TARPs) modulate AMPAR synaptic trafficking and transmission via disc-large (DLG) subfamily of membrane-associated guanylate kinases (MAGUKs). Despite extensive studies, the molecular mechanism governing specific TARP/MAGUK interaction remains elusive. Using stargazin and PSD-95 as the representatives, we discover that the entire tail of stargazin (Stg_CT) is required for binding to PSD-95. The PDZ binding motif (PBM) and an Arg-rich motif upstream of PBM conserved in TARPs bind to multiple sites on PSD-95, thus resulting in a highly specific and multivalent stargazin/PSD-95 complex. Stargazin in complex with PSD-95 or PSD-95-assembled postsynaptic complexes form highly concentrated and dynamic condensates via phase separation, reminiscent of stargazin/PSD-95-mediated AMPAR synaptic clustering and trapping. Importantly, charge neutralization mutations in TARP_CT Arg-rich motif weakened TARP's condensation with PSD-95 and impaired TARP-mediated AMPAR synaptic transmission in mice hippocampal neurons. The TARP_CT/PSD-95 interaction mode may have implications for understanding clustering of other synaptic transmembrane proteins.


Asunto(s)
Canales de Calcio/metabolismo , Homólogo 4 de la Proteína Discs Large/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Transmisión Sináptica , Animales , Guanilato-Quinasas/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Densidad Postsináptica/metabolismo , Transporte de Proteínas
8.
Nat Commun ; 9(1): 5205, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30510185

RESUMEN

The originally published version of this Article contained errors in Figure 5, for which we apologise. In panel c, the scatter graph was inadvertently replaced with a scatter graph comprising a subset of data points from panel d. Furthermore, the legends to Figures 5c and 5d were inverted. These errors have now been corrected in both the PDF and HTML versions of the Article, and the incorrect version of Fig. 5c is presented in the Author Correction associated with this Article.

9.
Nat Commun ; 9(1): 2069, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29802289

RESUMEN

CaMKII is one of the most studied synaptic proteins, but many critical issues regarding its role in synaptic function remain unresolved. Using a CRISPR-based system to delete CaMKII and replace it with mutated forms in single neurons, we have rigorously addressed its various synaptic roles. In brief, basal AMPAR and NMDAR synaptic transmission both require CaMKIIα, but not CaMKIIß, indicating that, even in the adult, synaptic transmission is determined by the ongoing action of CaMKIIα. While AMPAR transmission requires kinase activity, NMDAR transmission does not, implying a scaffolding role for the CaMKII protein instead. LTP is abolished in the absence of CaMKIIα and/or CaMKIIß and with an autophosphorylation impaired CaMKIIα (T286A). With the exception of NMDAR synaptic currents, all aspects of CaMKIIα signaling examined require binding to the NMDAR, emphasizing the essential role of this receptor as a master synaptic signaling hub.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hipocampo/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología , Animales , Sistemas CRISPR-Cas , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Membrana Celular/metabolismo , Femenino , Células HEK293 , Hipocampo/citología , Humanos , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/fisiología , Fosforilación , Ratas , Receptores de Glutamato/metabolismo , Transducción de Señal/fisiología , Sinapsis/fisiología
10.
Proc Natl Acad Sci U S A ; 115(23): E5373-E5381, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29784783

RESUMEN

The δ1 glutamate receptor (GluD1) was cloned decades ago and is widely expressed in many regions of the brain. However, its functional roles in these brain circuits remain unclear. Here, we find that GluD1 is required for both excitatory synapse formation and maintenance in the hippocampus. The action of GluD1 is absent in the Cbln2 knockout mouse. Furthermore, the GluD1 actions require the presence of presynaptic neurexin 1ß carrying the splice site 4 insert (+S4). Together, our findings demonstrate that hippocampal synapse assembly and maintenance require a tripartite molecular complex in which the ligand Cbln2 binds with presynaptic neurexin 1ß (+S4) and postsynaptic GluD1. We provide evidence that this mechanism may apply to other forebrain synapses, where GluD1 is widely expressed.


Asunto(s)
Hipocampo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Precursores de Proteínas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Glutamato/metabolismo , Sinapsis/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Proteínas de Unión al Calcio , Diferenciación Celular/fisiología , Células Cultivadas , Glutamato Deshidrogenasa , Hipocampo/citología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa/genética , Neuronas/citología , Neuronas/metabolismo , Precursores de Proteínas/genética , Ratas , Receptores de Superficie Celular/genética , Receptores de Glutamato/genética , Sinapsis/genética , Transmisión Sináptica
11.
Proc Natl Acad Sci U S A ; 115(15): 3948-3953, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29581259

RESUMEN

Long-term potentiation (LTP) is a persistent strengthening of synaptic transmission in the brain and is arguably the most compelling cellular and molecular model for learning and memory. Previous work found that both AMPA receptors and exogenously expressed kainate receptors are equally capable of expressing LTP, despite their limited homology and their association with distinct auxiliary subunits, indicating that LTP is far more promiscuous than previously thought. What might these two subtypes of glutamate receptor have in common? Using a single-cell molecular replacement strategy, we demonstrate that the AMPA receptor auxiliary subunit TARP γ-8, via its PDZ-binding motif, is indispensable for both basal synaptic transmission and LTP. Remarkably, kainate receptors and their auxiliary subunits Neto proteins share the same requirement of PDZ-binding domains for synaptic trafficking and LTP. Together, these results suggest that a minimal postsynaptic requirement for LTP is the PDZ binding of glutamate receptors/auxiliary subunits to PSD scaffolding proteins.


Asunto(s)
Canales de Calcio/metabolismo , Potenciación a Largo Plazo , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Animales , Canales de Calcio/química , Canales de Calcio/genética , Humanos , Dominios PDZ , Unión Proteica , Receptores AMPA/química , Receptores AMPA/genética , Receptores de Ácido Kaínico/genética , Receptores de Ácido Kaínico/metabolismo , Sinapsis/química , Sinapsis/genética
12.
Front Pharmacol ; 9: 1508, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30687088

RESUMEN

Alterations of the PI3K/Akt/mammalian target of rapamycin complex 1 (mTORC1) signaling pathway are causally involved in a subset of malformations of cortical development (MCDs) ranging from focal cortical dysplasia (FCD) to hemimegalencephaly and megalencephaly. These MCDs represent a frequent cause of refractory pediatric epilepsy. The endocannabinoid system -especially cannabinoid CB1 receptor- exerts a neurodevelopmental regulatory role at least in part via activation of mTORC1 signaling. Therefore, we sought to characterize the possible contribution of endocannabinoid system signaling to FCD. Confocal microscopy characterization of the CB1 receptor expression and mTORC1 activation was conducted in FCD Type II resection samples. FCD samples were subjected to single nucleotide polymorphism screening for endocannabinoid system elements, as well as CB1 receptor gene sequencing. Cannabinoid CB1 receptor levels were increased in FCD with overactive mTORC1 signaling. CB1 receptors were enriched in phospho-S6-positive cells including balloon cells (BCs) that co-express aberrant markers of undifferentiated cells and dysplastic neurons. Pharmacological regulation of CB1 receptors and the mTORC1 pathway was performed in fresh FCD-derived organotypic cultures. HU-210-evoked activation of CB1 receptors was unable to further activate mTORC1 signaling, whereas CB1 receptor blockade with rimonabant attenuated mTORC1 overactivation. Alterations of the endocannabinoid system may thus contribute to FCD pathological features, and blockade of cannabinoid signaling might be a new therapeutic intervention in FCD.

13.
Proc Natl Acad Sci U S A ; 114(50): 13266-13271, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29180434

RESUMEN

Bidirectional scaling of synaptic transmission, expressed as a compensatory change in quantal size following chronic activity perturbation, is a critical effector mechanism underlying homeostatic plasticity in the brain. An emerging model posits that the GluA2 AMPA receptor (AMPAR) subunit may be important for the bidirectional scaling of excitatory transmission; however, whether this subunit plays an obligatory role in synaptic scaling, and the identity of the precise domain(s) involved, remain controversial. We set out to determine the specific AMPAR subunit required for scaling up in CA1 hippocampal pyramidal neurons, and found that the GluA2 subunit is both necessary and sufficient. In addition, our results point to a critical role for a single amino acid within the membrane-proximal region of the GluA2 cytoplasmic tail, and suggest a distinct model for the regulation of AMPAR trafficking in synaptic homeostasis.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Receptores AMPA/metabolismo , Potenciales Sinápticos , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Homeostasis , Ratones , Dominios Proteicos , Transporte de Proteínas , Células Piramidales/metabolismo , Células Piramidales/fisiología , Receptores AMPA/química , Sinapsis/metabolismo , Sinapsis/fisiología
14.
Proc Natl Acad Sci U S A ; 114(27): 7136-7141, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28630296

RESUMEN

The amino-terminal domain (ATD) of AMPA receptors (AMPARs) accounts for approximately 50% of the protein, yet its functional role, if any, remains a mystery. We have discovered that the translocation of surface GluA1, but not GluA2, AMPAR subunits to the synapse requires the ATD. GluA1A2 heteromers in which the ATD of GluA1 is absent fail to translocate, establishing a critical role of the ATD of GluA1. Inserting GFP into the ATD interferes with the constitutive synaptic trafficking of GluA1, but not GluA2, mimicking the deletion of the ATD. Remarkably, long-term potentiation (LTP) can override the masking effect of the GFP tag. GluA1, but not GluA2, lacking the ATD fails to show LTP. These findings uncover a role for the ATD in subunit-specific synaptic trafficking of AMPARs, both constitutively and during plasticity. How LTP, induced postsynaptically, engages these extracellular trafficking motifs and what specific cleft proteins participate in the process remain to be elucidated.


Asunto(s)
Receptores AMPA/metabolismo , Sinapsis/metabolismo , Secuencias de Aminoácidos , Animales , Encéfalo/metabolismo , Citoplasma/metabolismo , Electroporación , Potenciales Postsinápticos Excitadores , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo , Ratones , Neuronas/metabolismo , Dominios Proteicos , Isoformas de Proteínas , Multimerización de Proteína , Ratas , Transmisión Sináptica
15.
Cereb Cortex ; 27(11): 5303-5317, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334226

RESUMEN

Neuronal migration is a fundamental process of brain development, and its disruption underlies devastating neurodevelopmental disorders. The transcriptional programs governing this process are relatively well characterized. However, how environmental cues instruct neuronal migration remains poorly understood. Here, we demonstrate that the cannabinoid CB1 receptor is strictly required for appropriate pyramidal neuron migration in the developing cortex. Acute silencing of the CB1 receptor alters neuronal morphology and impairs radial migration. Consequently, CB1 siRNA-electroporated mice display cortical malformations mimicking subcortical band heterotopias and increased seizure susceptibility in adulthood. Importantly, rescuing the CB1 deficiency-induced radial migration arrest by knockdown of the GTPase protein RhoA restored the hyperexcitable neuronal network and seizure susceptibility. Our findings show that CB1 receptor/RhoA signaling regulates pyramidal neuron migration, and that deficient CB1 receptor signaling may contribute to cortical development malformations leading to refractory epilepsy independently of its canonical neuromodulatory role in the adult brain.


Asunto(s)
Movimiento Celular/fisiología , Corteza Cerebral/anomalías , Corteza Cerebral/metabolismo , Células Piramidales/metabolismo , Receptor Cannabinoide CB1/deficiencia , Convulsiones/metabolismo , Animales , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/patología , Electroporación , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Hibridación in Situ , Ratones Transgénicos , Microscopía Confocal , Pentilenotetrazol , Células Piramidales/patología , ARN Interferente Pequeño , Receptor Cannabinoide CB1/genética , Convulsiones/patología , Técnicas de Cultivo de Tejidos , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
16.
Stroke ; 48(1): 204-212, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27899748

RESUMEN

BACKGROUND AND PURPOSE: Stroke is a leading cause of adult disability characterized by physical, cognitive, and emotional disturbances. Unfortunately, pharmacological options are scarce. The cannabinoid type-2 receptor (CB2R) is neuroprotective in acute experimental stroke by anti-inflammatory mechanisms. However, its role in chronic stroke is still unknown. METHODS: Stroke was induced by permanent middle cerebral artery occlusion in mice; CB2R modulation was assessed by administering the CB2R agonist JWH133 ((6aR,10aR)-3-(1,1-dimethylbutyl)-6a,7,10,10a-tetrahydro-6,6,9-trimethyl-6H-dibenzo[b,d]pyran) or the CB2R antagonist SR144528 (N-[(1S)-endo-1,3,3-trimethylbicyclo-[2.2.1]-heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide) once daily from day 3 to the end of the experiment or by CB2R genetic deletion. Analysis of immunofluorescence-labeled brain sections, 5-bromo-2´-deoxyuridine (BrdU) staining, fluorescence-activated cell sorter analysis of brain cell suspensions, and behavioral tests were performed. RESULTS: SR144528 decreased neuroblast migration toward the boundary of the infarct area when compared with vehicle-treated mice 14 days after middle cerebral artery occlusion. Consistently, mice on this pharmacological treatment, like mice with CB2R genetic deletion, displayed a lower number of new neurons (NeuN+/BrdU+ cells) in peri-infarct cortex 28 days after stroke when compared with vehicle-treated group, an effect accompanied by a worse sensorimotor performance in behavioral tests. The CB2R agonist did not affect neurogenesis or outcome in vivo, but increased the migration of neural progenitor cells in vitro; the CB2R antagonist alone did not affect in vitro migration. CONCLUSIONS: Our data support that CB2R is fundamental for driving neuroblast migration and suggest that an endocannabinoid tone is required for poststroke neurogenesis by promoting neuroblast migration toward the injured brain tissue, increasing the number of new cortical neurons and, conceivably, enhancing motor functional recovery after stroke.


Asunto(s)
Neurogénesis/fisiología , Receptor Cannabinoide CB2/fisiología , Recuperación de la Función/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Canfanos/farmacología , Cannabinoides/farmacología , Cannabinoides/uso terapéutico , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Pirazoles/farmacología , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/antagonistas & inhibidores , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Resultado del Tratamiento
17.
Hum Reprod ; 32(1): 175-184, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27821707

RESUMEN

STUDY QUESTION: Does signaling via the cannabinoid (CB1) receptor play a role in the pathogenesis of endometriosis in a mouse model? SUMMARY ANSWER: Mice treated with a CB1 agonist developed larger ectopic lesions, while less severe lesions developed in the absence of functional CB1 expression. WHAT IS KNOWN ALREADY: The expression of components of the endocannabinoid system has been demonstrated in both mouse and human uteri. CB1 receptors are expressed in human epithelial and stromal cell lines derived from eutopic endometrium and deep infiltrating endometriosis nodules. STUDY DESIGN, SIZE, DURATION: This was a randomized study in a mouse model of endometriosis. In a first set of experiments, mice with endometriosis were treated with the CB1 receptor agonist methanandamide (MET) (5 mg/kg, n = 20) on Days 1-5 and 8-12. In a second set of experiments, endometriosis development was evaluated in CB1-/- mice and in their wild-type (WT) littermates. PARTICIPANTS/MATERIALS, SETTING, METHODS: Endometriosis-like lesions were induced in Balb/c and C57/Bl6 mice. Two weeks after disease induction, the lesions were counted, measured and either included for immunohistochemistry analysis or frozen for gene expression profiling by semi-quantitative real-time PCR. To limit the role of chance, the experiments were conducted under standardized laboratory conditions with appropriate controls. MAIN RESULTS AND THE ROLE OF CHANCE: The lesion total volume was significantly higher in MET-treated compared with vehicle-treated mice (P < 0.05). Expression levels of mRNA for survivin, N-cadherin, integrin ß1 and interleukin-6 were increased in the ectopic endometrium of MET-treated versus vehicle-treated mice (P < 0.05). CB1-/- recipients that received endometrial tissue fragments from CB1-/- donors, WT recipients that received endometrial tissue fragments from CB1-/- donors and CB1-/- recipients that received endometrial tissue fragments from WT donors all showed a significant reduction in total lesion volume and lower expression of survivin and N-cadherin compared with WT recipients receiving uterine fragments from WT donors (P < 0.05). LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: We provide evidence that endocannabinoid signaling via CB1 receptor plays a role in the development of endometriosis in a mouse model. However, the relative contribution of the CB1-mediated signaling pathways active in inflammatory, uterine and peritoneal cells remains to be ascertained. Since the study was performed in a mouse model, the significance of the findings in the human system warrants further investigation. WIDER IMPLICATIONS OF THE FINDINGS: Clarifying the function and regulation of CB1 and its molecular interactions with endogenous ligands, and how endocannabinoids levels are regulated in women with endometriosis, represent critical areas of research for the potential development of a novel medical treatment of the disease. STUDY FUNDING/COMPETING INTERESTS: A.M.S. was supported by a fellowship from Fondazione Giorgio Pardi. The authors have no conflicts of interest to declare.


Asunto(s)
Ácidos Araquidónicos/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Endometriosis/patología , Enfermedades Peritoneales/patología , Receptor Cannabinoide CB1/metabolismo , Animales , Cadherinas/metabolismo , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Endometriosis/metabolismo , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Endometrio/patología , Femenino , Proteínas Inhibidoras de la Apoptosis/metabolismo , Integrina beta1/metabolismo , Interleucina-6/metabolismo , Ratones , Enfermedades Peritoneales/metabolismo , Peritoneo/efectos de los fármacos , Peritoneo/metabolismo , Peritoneo/patología , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Survivin
18.
Sci Rep ; 6: 29789, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27430371

RESUMEN

Cannabinoids have shown to exert neuroprotective actions in animal models by acting at different targets including canonical cannabinoid receptors and PPARγ. We previously showed that VCE-003, a cannabigerol (CBG) quinone derivative, is a novel neuroprotective and anti-inflammatory cannabinoid acting through PPARγ. We have now generated a non-thiophilic VCE-003 derivative named VCE-003.2 that preserves the ability to activate PPARγ and analyzed its neuroprotective activity. This compound exerted a prosurvival action in progenitor cells during neuronal differentiation, which was prevented by a PPARγ antagonist, without affecting neural progenitor cell proliferation. In addition, VCE-003.2 attenuated quinolinic acid (QA)-induced cell death and caspase-3 activation and also reduced mutant huntingtin aggregates in striatal cells. The neuroprotective profile of VCE-003.2 was analyzed using in vivo models of striatal neurodegeneration induced by QA and 3-nitropropionic acid (3NP) administration. VCE-003.2 prevented medium spiny DARPP32(+) neuronal loss in these Huntington's-like disease mice models improving motor deficits, reactive astrogliosis and microglial activation. In the 3NP model VCE-003.2 inhibited the upregulation of proinflammatory markers and improved antioxidant defenses in the brain. These data lead us to consider VCE-003.2 to have high potential for the treatment of Huntington's disease (HD) and other neurodegenerative diseases with neuroinflammatory traits.


Asunto(s)
Cannabinoides/farmacología , Modelos Animales de Enfermedad , Enfermedad de Huntington/prevención & control , Células-Madre Neurales/efectos de los fármacos , Quinonas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Enfermedad de Huntington/patología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Células-Madre Neurales/fisiología , Fármacos Neuroprotectores/farmacología , Ratas
19.
Proc Natl Acad Sci U S A ; 112(44): 13693-8, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26460022

RESUMEN

The CB1 cannabinoid receptor, the main target of Δ(9)-tetrahydrocannabinol (THC), the most prominent psychoactive compound of marijuana, plays a crucial regulatory role in brain development as evidenced by the neurodevelopmental consequences of its manipulation in animal models. Likewise, recreational cannabis use during pregnancy affects brain structure and function of the progeny. However, the precise neurobiological substrates underlying the consequences of prenatal THC exposure remain unknown. As CB1 signaling is known to modulate long-range corticofugal connectivity, we analyzed the impact of THC exposure on cortical projection neuron development. THC administration to pregnant mice in a restricted time window interfered with subcerebral projection neuron generation, thereby altering corticospinal connectivity, and produced long-lasting alterations in the fine motor performance of the adult offspring. Consequences of THC exposure were reminiscent of those elicited by CB1 receptor genetic ablation, and CB1-null mice were resistant to THC-induced alterations. The identity of embryonic THC neuronal targets was determined by a Cre-mediated, lineage-specific, CB1 expression-rescue strategy in a CB1-null background. Early and selective CB1 reexpression in dorsal telencephalic glutamatergic neurons but not forebrain GABAergic neurons rescued the deficits in corticospinal motor neuron development of CB1-null mice and restored susceptibility to THC-induced motor alterations. In addition, THC administration induced an increase in seizure susceptibility that was mediated by its interference with CB1-dependent regulation of both glutamatergic and GABAergic neuron development. These findings demonstrate that prenatal exposure to THC has long-lasting deleterious consequences in the adult offspring solely mediated by its ability to disrupt the neurodevelopmental role of CB1 signaling.


Asunto(s)
Corteza Cerebral/metabolismo , Dronabinol/administración & dosificación , Exposición Materna , Neuronas/metabolismo , Receptor Cannabinoide CB1/metabolismo , Animales , Corteza Cerebral/crecimiento & desarrollo , Femenino , Ratones , Embarazo
20.
Nat Commun ; 6: 6474, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25753651

RESUMEN

The proneural factor Ascl1 controls multiple steps of neurogenesis in the embryonic brain, including progenitor division and neuronal migration. Here we show that Cenpj, also known as CPAP, a microcephaly gene, is a transcriptional target of Ascl1 in the embryonic cerebral cortex. We have characterized the role of Cenpj during cortical development by in utero electroporation knockdown and found that silencing Cenpj in the ventricular zone disrupts centrosome biogenesis and randomizes the cleavage plane orientation of radial glia progenitors. Moreover, we show that downregulation of Cenpj in post-mitotic neurons increases stable microtubules and leads to slower neuronal migration, abnormal centrosome position and aberrant neuronal morphology. Moreover, rescue experiments shows that Cenpj mediates the role of Ascl1 in centrosome biogenesis in progenitor cells and in microtubule dynamics in migrating neurons. These data provide insights into genetic pathways controlling cortical development and primary microcephaly observed in humans with mutations in Cenpj.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Corteza Cerebral/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , División Celular , Movimiento Celular , Centrosoma/metabolismo , Centrosoma/ultraestructura , Corteza Cerebral/citología , Electroporación , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Inyecciones Intraventriculares , Ratones , Ratones Transgénicos , Microtomía , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Células-Madre Neurales/ultraestructura , Neuronas/ultraestructura , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA