Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 15(706): eadd1014, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37494470

RESUMEN

Optogenetics is a widely used technology with potential for translational research. A critical component of such applications is the ability to track the location of the transduced opsin in vivo. To address this problem, we engineered an excitatory opsin, ChRERα (hChR2(134R)-V5-ERα-LBD), that could be visualized using positron emission tomography (PET) imaging in a noninvasive, longitudinal, and quantitative manner. ChRERα consists of the prototypical excitatory opsin channelrhodopsin-2 (ChR2) and the ligand-binding domain (LBD) of the human estrogen receptor α (ERα). ChRERα showed conserved ChR2 functionality and high affinity for [18F]16α-fluoroestradiol (FES), an FDA-approved PET radiopharmaceutical. Experiments in rats demonstrated that adeno-associated virus (AAV)-mediated expression of ChRERα enables neural circuit manipulation in vivo and that ChRERα expression could be monitored using FES-PET imaging. In vivo experiments in nonhuman primates (NHPs) confirmed that ChRERα expression could be monitored at the site of AAV injection in the primary motor cortex and in long-range neuronal terminals for up to 80 weeks. The anatomical connectivity map of the primary motor cortex identified by FES-PET imaging of ChRERα expression overlapped with a functional connectivity map identified using resting state fMRI in a separate cohort of NHPs. Overall, our results demonstrate that ChRERα expression can be mapped longitudinally in the mammalian brain using FES-PET imaging and can be used for neural circuit modulation in vivo.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno , Ratas , Humanos , Animales , Femenino , Receptor alfa de Estrógeno/metabolismo , Opsinas/metabolismo , Tomografía de Emisión de Positrones , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Primates , Estradiol/metabolismo , Neoplasias de la Mama/metabolismo , Mamíferos/metabolismo
2.
bioRxiv ; 2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37333325

RESUMEN

Ketamine's role in providing a rapid and sustained antidepressant response, particularly for patients unresponsive to conventional treatments, is increasingly recognized. A core symptom of depression, anhedonia, or the loss of enjoyment or interest in previously pleasurable activities, is known to be significantly alleviated by ketamine. While several hypotheses have been proposed regarding the mechanisms by which ketamine alleviates anhedonia, the specific circuits and synaptic changes responsible for its sustained therapeutic effects are not yet understood. Here, we show that the nucleus accumbens (NAc), a major hub of the reward circuitry, is essential for ketamine's effect in rescuing anhedonia in mice subjected to chronic stress, a critical risk factor in the genesis of depression in humans. Specifically, a single exposure to ketamine rescues stress-induced decreased strength of excitatory synapses on NAc D1 dopamine receptor-expressing medium spiny neurons (D1-MSNs). By using a novel cell-specific pharmacology method, we demonstrate that this cell-type specific neuroadaptation is necessary for the sustained therapeutic effects of ketamine. To test for causal sufficiency, we artificially mimicked ketamine-induced increase in excitatory strength on D1-MSNs and found that this recapitulates the behavioral amelioration induced by ketamine. Finally, to determine the presynaptic origin of the relevant glutamatergic inputs for ketamine-elicited synaptic and behavioral effects, we used a combination of opto- and chemogenetics. We found that ketamine rescues stress-induced reduction in excitatory strength at medial prefrontal cortex and ventral hippocampus inputs to NAc D1-MSNs. Chemogenetically preventing ketamine-evoked plasticity at those unique inputs to the NAc reveals a ketamine-operated input-specific control of hedonic behavior. These results establish that ketamine rescues stress-induced anhedonia via cell-type-specific adaptations as well as information integration in the NAc via discrete excitatory synapses.

3.
Transl Psychiatry ; 11(1): 570, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34750356

RESUMEN

Cocaine binds to the dopamine (DA) transporter (DAT) to regulate cocaine reward and seeking behavior. Zinc (Zn2+) also binds to the DAT, but the in vivo relevance of this interaction is unknown. We found that Zn2+ concentrations in postmortem brain (caudate) tissue from humans who died of cocaine overdose were significantly lower than in control subjects. Moreover, the level of striatal Zn2+ content in these subjects negatively correlated with plasma levels of benzoylecgonine, a cocaine metabolite indicative of recent use. In mice, repeated cocaine exposure increased synaptic Zn2+ concentrations in the caudate putamen (CPu) and nucleus accumbens (NAc). Cocaine-induced increases in Zn2+ were dependent on the Zn2+ transporter 3 (ZnT3), a neuronal Zn2+ transporter localized to synaptic vesicle membranes, as ZnT3 knockout (KO) mice were insensitive to cocaine-induced increases in striatal Zn2+. ZnT3 KO mice showed significantly lower electrically evoked DA release and greater DA clearance when exposed to cocaine compared to controls. ZnT3 KO mice also displayed significant reductions in cocaine locomotor sensitization, conditioned place preference (CPP), self-administration, and reinstatement compared to control mice and were insensitive to cocaine-induced increases in striatal DAT binding. Finally, dietary Zn2+ deficiency in mice resulted in decreased striatal Zn2+ content, cocaine locomotor sensitization, CPP, and striatal DAT binding. These results indicate that cocaine increases synaptic Zn2+ release and turnover/metabolism in the striatum, and that synaptically released Zn2+ potentiates the effects of cocaine on striatal DA neurotransmission and behavior and is required for cocaine-primed reinstatement. In sum, these findings reveal new insights into cocaine's pharmacological mechanism of action and suggest that Zn2+ may serve as an environmentally derived regulator of DA neurotransmission, cocaine pharmacodynamics, and vulnerability to cocaine use disorders.


Asunto(s)
Cocaína , Dopamina , Animales , Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ratones , Núcleo Accumbens/metabolismo , Transmisión Sináptica , Zinc
4.
Immunity ; 54(2): 225-234.e6, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33476547

RESUMEN

Microglia are activated in many neurological diseases and have been suggested to play an important role in the development of affective disorders including major depression. To investigate how microglial signaling regulates mood, we used bidirectional chemogenetic manipulations of microglial activity in mice. Activation of microglia in the dorsal striatum induced local cytokine expression and a negative affective state characterized by anhedonia and aversion, whereas inactivation of microglia blocked aversion induced by systemic inflammation. Interleukin-6 signaling and cyclooxygenase-1 mediated prostaglandin synthesis in the microglia were critical for the inflammation-induced aversion. Correspondingly, microglial activation led to a prostaglandin-dependent reduction of the excitability of striatal neurons. These findings demonstrate a mechanism by which microglial activation causes negative affect through prostaglandin-dependent modulation of striatal neurons and indicate that interference with this mechanism could milden the depressive symptoms in somatic and psychiatric diseases involving microglial activation.


Asunto(s)
Anhedonia/fisiología , Cuerpo Estriado/inmunología , Depresión/inmunología , Microglía/inmunología , Neuronas/fisiología , Animales , Animales Modificados Genéticamente , Conducta Animal , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inflamación , Interleucina-6/metabolismo , Activación de Macrófagos , Ratones , Inflamación Neurogénica , Prostaglandinas/metabolismo
5.
Mol Psychiatry ; 26(6): 1860-1879, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32161361

RESUMEN

Stress promotes negative affective states, which include anhedonia and passive coping. While these features are in part mediated by neuroadaptations in brain reward circuitry, a comprehensive framework of how stress-induced negative affect may be encoded within key nodes of this circuit is lacking. Here, we show in a mouse model for stress-induced anhedonia and passive coping that these phenomena are associated with increased synaptic strength of ventral hippocampus (VH) excitatory synapses onto D1 medium spiny neurons (D1-MSNs) in the nucleus accumbens medial shell (NAcmSh), and with lateral hypothalamus (LH)-projecting D1-MSN hyperexcitability mediated by decreased inwardly rectifying potassium channel (IRK) function. Stress-induced negative affective states are prevented by depotentiation of VH to NAcmSh synapses, restoring Kir2.1 function in D1R-MSNs, or disrupting co-participation of these synaptic and intrinsic adaptations in D1-MSNs. In conclusion, our data provide strong evidence for a disynaptic pathway controlling maladaptive emotional behavior.


Asunto(s)
Anhedonia , Receptores de Dopamina D1 , Adaptación Psicológica , Animales , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(15): 8611-8615, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32229573

RESUMEN

Electrical or optogenetic stimulation of lateral hypothalamic (LH) GABA neurons induces rapid vigorous eating in sated animals. The dopamine system has been implicated in the regulation of feeding. Previous work has suggested that a subset of LH GABA neurons projects to the ventral tegmental area (VTA) and targets GABA neurons, inhibiting them and thereby disinhibiting dopaminergic activity and release. Furthermore, stimulation-induced eating is attenuated by dopamine lesions or receptor antagonists. Here we explored the involvement of dopamine in LH stimulation-induced eating. LH stimulation caused sated mice to pick up pellets of standard chow with latencies that varied based on stimulation intensity; once food was picked up, animals ate for the remainder of the 60-s stimulation period. However, lesion of VTA GABA neurons failed to disrupt this effect. Moreover, direct stimulation of VTA or substantia nigra dopamine cell bodies failed to induce food approach or eating. Looking further, we found that some LH GABA fibers pass through the VTA to more caudal sites, where they synapse onto neurons near the locus coeruleus (LC). Similar eating was induced by stimulation of LH GABA terminals or GABA cell bodies in this peri-LC region. Lesion of peri-LC GABA neurons blocked LH stimulation-induced eating, establishing them as a critical downstream circuit element for LH neurons. Surprisingly, lesions did not alter body weight, suggesting that this system is not involved in the hunger or satiety mechanisms that govern normal feeding. Thus, we present a characterization of brain circuitry that may promote overeating and contribute to obesity.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Neuronas GABAérgicas/metabolismo , Área Hipotalámica Lateral/fisiología , Área Tegmental Ventral/fisiología , Animales , Conducta Animal , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Femenino , Neuronas GABAérgicas/citología , Área Hipotalámica Lateral/citología , Masculino , Ratones , Vías Nerviosas , Receptores de GABA-A/metabolismo , Recompensa , Área Tegmental Ventral/citología , Ácido gamma-Aminobutírico/metabolismo
7.
Neuron ; 97(2): 261-262, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29346748

RESUMEN

How do nucleus accumbens (NAc) subdivisions shape information flow into distinct ventral tegmental area (VTA) subcircuits? Yang et al. (2018) provide insightful answers to this question by expanding our knowledge about the circuit architecture and function of reciprocal connectivity between NAc and VTA.


Asunto(s)
Núcleo Accumbens , Área Tegmental Ventral , Dopamina
8.
Sci Transl Med ; 9(420)2017 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-29237760

RESUMEN

The AAA+ adenosine triphosphatase (ATPase) Thorase plays a critical role in controlling synaptic plasticity by regulating the expression of surface α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs). Bidirectional sequencing of exons of ATAD1, the gene encoding Thorase, in a cohort of patients with schizophrenia and healthy controls revealed rare Thorase variants. These variants caused defects in glutamatergic signaling by impairing AMPAR internalization and recycling in mouse primary cortical neurons. This contributed to increased surface expression of the AMPAR subunit GluA2 and enhanced synaptic transmission. Heterozygous Thorase-deficient mice engineered to express these Thorase variants showed altered synaptic transmission and several behavioral deficits compared to heterozygous Thorase-deficient mice expressing wild-type Thorase. These behavioral impairments were rescued by the competitive AMPAR antagonist Perampanel, a U.S. Food and Drug Administration-approved drug. These findings suggest that Perampanel may be useful for treating disorders involving compromised AMPAR-mediated glutamatergic neurotransmission.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Variación Genética , Glutamatos/metabolismo , Piridonas/farmacología , Transmisión Sináptica/efectos de los fármacos , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Conducta Animal , Células Cultivadas , Corteza Cerebral/patología , Endocitosis/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Heterocigoto , Humanos , Memoria/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nitrilos , Multimerización de Proteína , Conducta Social
10.
Nat Neurosci ; 20(3): 438-448, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28114294

RESUMEN

Afferent inputs to the ventral tegmental area (VTA) control reward-related behaviors through regulation of dopamine neuron activity. The nucleus accumbens (NAc) provides one of the most prominent projections to the VTA; however, recent studies have provided conflicting evidence regarding the function of these inhibitory inputs. Using optogenetics, cell-specific ablation, whole cell patch-clamp and immuno-electron microscopy, we found that NAc inputs synapsed directly onto dopamine neurons, preferentially activating GABAB receptors. GABAergic inputs from the NAc and local VTA GABA neurons were differentially modulated and activated separate receptor populations in dopamine neurons. Genetic deletion of GABAB receptors from dopamine neurons in adult mice did not affect general or morphine-induced locomotor activity, but markedly increased cocaine-induced locomotion. Collectively, our findings demonstrate notable selectivity in the inhibitory architecture of the VTA and suggest that long-range GABAergic inputs to dopamine neurons fundamentally regulate behavioral responses to cocaine.


Asunto(s)
Cocaína/farmacología , Inhibición Neural/fisiología , Núcleo Accumbens/fisiología , Receptores de GABA-B/fisiología , Recompensa , Área Tegmental Ventral/fisiología , Animales , Neuronas Dopaminérgicas/fisiología , Neuronas Dopaminérgicas/ultraestructura , Femenino , Técnicas de Silenciamiento del Gen , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Morfina/farmacología , Receptor de Adenosina A1/fisiología , Receptores de GABA-A/fisiología , Receptores de GABA-B/biosíntesis , Receptores de GABA-B/genética , Transmisión Sináptica/fisiología , Área Tegmental Ventral/ultraestructura
11.
Neuron ; 93(2): 425-440, 2017 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-28103482

RESUMEN

Fear learning is a fundamental behavioral process that requires dopamine (DA) release. Experience-dependent synaptic plasticity occurs on DA neurons while an organism is engaged in aversive experiences. However, whether synaptic plasticity onto DA neurons is causally involved in aversion learning is unknown. Here, we show that a stress priming procedure enhances fear learning by engaging VTA synaptic plasticity. Moreover, we took advantage of the ability of the ATPase Thorase to regulate the internalization of AMPA receptors (AMPARs) in order to selectively manipulate glutamatergic synaptic plasticity on DA neurons. Genetic ablation of Thorase in DAT+ neurons produced increased AMPAR surface expression and function that lead to impaired induction of both long-term depression (LTD) and long-term potentiation (LTP). Strikingly, animals lacking Thorase in DAT+ neurons expressed greater associative learning in a fear conditioning paradigm. In conclusion, our data provide a novel, causal link between synaptic plasticity onto DA neurons and fear learning.


Asunto(s)
Aprendizaje por Asociación/fisiología , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Miedo , Plasticidad Neuronal/fisiología , Estrés Psicológico , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Conducta Animal , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Técnicas de Inactivación de Genes , Captura por Microdisección con Láser , Aprendizaje/fisiología , Potenciación a Largo Plazo/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Ratones , Técnicas de Placa-Clamp , Receptores AMPA/metabolismo , Área Tegmental Ventral
12.
Neuron ; 93(1): 147-163, 2017 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-28056342

RESUMEN

Endogenous dynorphin signaling via the kappa-opioid receptor (KOR) in the nucleus accumbens (NAcc) powerfully mediates negative affective states and stress reactivity. Excitatory inputs from the hippocampus and amygdala play a fundamental role in shaping the activity of both NAcc D1 and D2 MSNs, which encode positive and negative motivational valences, respectively. However, a circuit-based mechanism by which KOR modulation of excitation-inhibition balance modifies D1 and D2 MSN activity is lacking. Here, we provide a comprehensive synaptic framework wherein presynaptic KOR inhibition decreases the excitatory drive of D1 MSN activity by the amygdala, but not the hippocampus. Conversely, presynaptic inhibition by KORs of inhibitory synapses on D2 MSNs enhances integration of excitatory drive by the amygdala and hippocampus. In conclusion, we describe a circuit-based mechanism showing differential gating of afferent control of D1 and D2 MSN activity by KORs in a pathway-specific manner.


Asunto(s)
Afecto/fisiología , Amígdala del Cerebelo/metabolismo , Dinorfinas/metabolismo , Hipocampo/metabolismo , Inhibición Neural/fisiología , Neuronas/metabolismo , Núcleo Accumbens/metabolismo , Receptores Opioides kappa/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Dinorfinas/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Hipocampo/fisiología , Masculino , Ratones , Motivación , Neuronas/fisiología , Núcleo Accumbens/fisiología , Técnicas de Placa-Clamp , Receptores Opioides kappa/genética , Receptores Opioides kappa/fisiología
13.
Neuron ; 86(5): 1145-57, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26050034

RESUMEN

The brain is wired to predict future outcomes. Experience-dependent plasticity at excitatory synapses within dopamine neurons of the ventral tegmental area, a key region for a broad range of motivated behaviors, is thought to be a fundamental cellular mechanism that enables adaptation to a dynamic environment. Thus, depending on the circumstances, dopamine neurons are capable of processing both positive and negative reinforcement learning strategies. In this review, we will discuss how changes in synaptic plasticity of dopamine neurons may affect dopamine release, as well as behavioral adaptations to different environmental conditions falling at opposite ends of a saliency spectrum ranging from reward to aversion.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Plasticidad Neuronal/fisiología , Refuerzo en Psicología , Recompensa , Sinapsis/fisiología , Animales , Dopamina/fisiología , Humanos
14.
J Neurosci ; 34(13): 4558-66, 2014 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-24672001

RESUMEN

Angelman syndrome (AS) is caused by the loss of Ube3A, an ubiquitin ligase that commits specific proteins to proteasomal degradation. How this defect causes autism and other pathological phenotypes associated with AS is unknown. Long-term depression (LTD) of excitatory synaptic transmission mediated by type 5 metabotropic glutamate (mGlu5) receptors was enhanced in hippocampal slices of Ube3A(m-/p+) mice, which model AS. No changes were found in NMDA-dependent LTD induced by low-frequency stimulation. mGlu5 receptor-dependent LTD in AS mice was sensitive to the protein synthesis inhibitor anisomycin, and relied on the same signaling pathways as in wild-type mice, e.g., the mitogen-activated protein kinase (MAPK) pathway, the phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycine pathway, and protein tyrosine phosphatase. Neither the stimulation of MAPK and PI3K nor the increase in Arc (activity-regulated cytoskeleton-associated protein) levels in response to mGlu5 receptor activation were abnormal in hippocampal slices from AS mice compared with wild-type mice. mGlu5 receptor expression and mGlu1/5 receptor-mediated polyphosphoinositide hydrolysis were also unchanged in the hippocampus of AS mice. In contrast, AS mice showed a reduced expression of the short Homer protein isoform Homer 1a, and an increased coupling of mGlu5 receptors to Homer 1b/c proteins in the hippocampus. These findings support the link between Homer proteins and monogenic autism, and lay the groundwork for the use of mGlu5 receptor antagonists in AS.


Asunto(s)
Síndrome de Angelman/genética , Síndrome de Angelman/patología , Proteínas Portadoras/metabolismo , Hipocampo/fisiopatología , Depresión Sináptica a Largo Plazo/fisiología , Receptor del Glutamato Metabotropico 5/metabolismo , Ubiquitina-Proteína Ligasas/genética , Animales , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Hemicigoto , Hipocampo/patología , Proteínas de Andamiaje Homer , Inmunosupresores/farmacología , Técnicas In Vitro , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirolimus/farmacología
15.
Biol Psychiatry ; 75(9): 701-10, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23541633

RESUMEN

BACKGROUND: Parkinson's disease (PD) is characterized by the progressive degeneration of the nigrostriatal dopaminergic pathway and the emergence of rigidity, tremor, and bradykinesia. Accumulating evidence indicates that PD is also accompanied by nonmotor symptoms including cognitive deficits, often manifested as impaired visuospatial memory. METHODS: We studied cognitive performance and synaptic plasticity in a mouse model of PD, characterized by partial lesion of the dopaminergic and noradrenergic inputs to striatum and hippocampus. Sham- and 6-hydroxydopamine-lesioned mice were subjected to the novel object recognition test, and long-term potentiation was examined in the dentate gyrus and CA1 regions of the hippocampus. RESULTS: Bilateral 6-hydroxydopamine lesion reduced long-term but not short-term novel object recognition and decreased long-term potentiation specifically in the dentate gyrus. These abnormalities did not depend on the loss of noradrenaline but were abolished by the antiparkinsonian drug, L-DOPA, or by SKF81297, a dopamine D1-type receptor agonist. In contrast, activation of dopamine D2-type receptors did not modify the effects produced by the lesion. Blockade of the extracellular signal-regulated kinases prevented the ability of SKF81297 to rescue novel object recognition and long-term potentiation. CONCLUSIONS: These findings show that partial dopamine depletion leads to impairment of long-term recognition memory accompanied by abnormal synaptic plasticity in the dentate gyrus. They also demonstrate that activation of dopamine D1 receptors corrects these deficits, through a mechanism that requires intact extracellular signal-regulated kinases signaling.


Asunto(s)
Trastornos del Conocimiento/fisiopatología , Giro Dentado/fisiopatología , Potenciación a Largo Plazo/fisiología , Trastornos Parkinsonianos/fisiopatología , Reconocimiento en Psicología/fisiología , Animales , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/fisiopatología , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/fisiopatología , Giro Dentado/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones Endogámicos C57BL , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiopatología , Oxidopamina , Trastornos Parkinsonianos/complicaciones , Trastornos Parkinsonianos/tratamiento farmacológico , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Factores de Tiempo
16.
Curr Pharm Des ; 19(36): 6480-90, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23432715

RESUMEN

The term "Autism Spectrum" is often used to describe disorders that are currently classified as Pervasive Developmental Disorders. These disorders are typically characterized by social deficits, communication difficulties, stereotyped or repetitive behaviors and/or cognitive delays or mental retardation; sometimes they present high comorbidity rates with epilepsy. Although these diagnoses share some common features, individuals with these disorders are thought to be "on the spectrum" because of differences in severity across these domains. Recent advances in the genetics of autism spectrum disorders (ASDs) are offering new valuable insights into molecular and cellular mechanisms of pathology. Of particular interest are transgenic technologies that allowed the engineering of several mouse models mimicking different kinds of monogenic heritable forms of ASDs. These transgenic models provide excellent opportunities to explore in detail cellular and molecular mechanisms underlying disease pathology and to identify novel targets for therapeutic intervention. Increasing evidence suggests that the pathophysiological core of the murine model is primarily due to changes in normal synaptic transmission and plasticity. Here, we will extensively review the synaptic alterations across different animal models of ASDs and recapitulate the pharmacological strategies aimed at rescuing hippocampal plasticity phenotypes. We describe how pharmacological modulation of mGlu5 receptor, through the use of positive or negative allosteric modulators (depending on the specific disorder), may represent a promising therapeutic strategy for ASDs treatment.


Asunto(s)
Antipsicóticos/uso terapéutico , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/terapia , Predisposición Genética a la Enfermedad , Plasticidad Neuronal/genética , Sinapsis/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores , Sinapsis/patología
17.
PLoS One ; 8(1): e54666, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23355887

RESUMEN

Abnormal use-dependent synaptic plasticity is universally accepted as the main physiological correlate of memory deficits in neurodegenerative disorders. It is unclear whether synaptic plasticity deficits take place during neuroinflammatory diseases, such as multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis (EAE). In EAE mice, we found significant alterations of synaptic plasticity rules in the hippocampus. When compared to control mice, in fact, hippocampal long-term potentiation (LTP) induction was favored over long-term depression (LTD) in EAE, as shown by a significant rightward shift in the frequency-synaptic response function. Notably, LTP induction was also enhanced in hippocampal slices from control mice following interleukin-1ß (IL-1ß) perfusion, and both EAE and IL-1ß inhibited GABAergic spontaneous inhibitory postsynaptic currents (sIPSC) without affecting glutamatergic transmission and AMPA/NMDA ratio. EAE was also associated with selective loss of GABAergic interneurons and with reduced gamma-frequency oscillations in the CA1 region of the hippocampus. Finally, we provided evidence that microglial activation in the EAE hippocampus was associated with IL-1ß expression, and hippocampal slices from control mice incubated with activated microglia displayed alterations of GABAergic transmission similar to those seen in EAE brains, through a mechanism dependent on enhanced IL-1ß signaling. These data may yield novel insights into the basis of cognitive deficits in EAE and possibly of MS.


Asunto(s)
Región CA1 Hipocampal , Encefalomielitis Autoinmune Experimental , Potenciación a Largo Plazo , Esclerosis Múltiple , Sinapsis , Transmisión Sináptica , Animales , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Línea Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Femenino , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/patología , Interleucina-1beta/metabolismo , Ratones , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , N-Metilaspartato/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo
18.
Neuropharmacology ; 66: 339-47, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22709946

RESUMEN

Alterations of the glutamatergic system have been implicated in the pathophysiology and treatment of major depression. In order to investigate the expression and function of mGlu5 receptors in an animal model for treatment-resistant depression we used rats bred for congenital learned helplessness (cLH) and the control strain, bred for resistance against inescapable stress, congenitally. not learned helpless rats (cNLH). Western blot analysis showed an increased expression of mGlu5 (but not mGlu1a) receptors in the hippocampus of cLH rats, as compared with control cNLH rats. We also examined mGlu1/5 receptor signaling by in vivo measurement of DHPG-stimulated polyphosphoinositides hydrolysis. Stimulation of (3)H-inositolmonophosphate formation induced by i.c.v. injection of DHPG was enhanced by about 50% in the hippocampus of cLH rats. Correspondingly, DHPG-induced long-term depression (LTD) at Schaffer collateral/CA1 pyramidal cell synapses was amplified in hippocampal slices of cLH rats, whereas LTD induced by low frequency stimulation of the Schaffer collaterals did not change. Moreover, these effects were associated with decreased basal dendritic spine density of CA1 pyramidal cell in cLH rats. These data raise the attractive possibility that changes in the expression and function of mGlu5 receptors in the hippocampus might underlie the changes in synaptic plasticity associated with the depressive-like phenotype of cLH rats. However, chronic treatment of cLH rats with MPEP did not reverse learned helplessness, indicating that the enhanced mGlu5 receptor function is not the only player in the behavioral phenotype of this genetic model of depression. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.


Asunto(s)
Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología , Desamparo Adquirido , Depresión Sináptica a Largo Plazo/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Sinapsis/fisiología , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Región CA3 Hipocampal/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Estimulación Eléctrica/métodos , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Hidrólisis/efectos de los fármacos , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Masculino , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Fosfatos de Fosfatidilinositol/metabolismo , Células Piramidales/citología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/biosíntesis , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
19.
Mol Neurobiol ; 46(3): 572-87, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22914888

RESUMEN

In the past years, major efforts have been made to understand the genetics and molecular pathogenesis of Alzheimer's disease (AD), which has been translated into extensive experimental approaches aimed at slowing down or halting disease progression. Advances in transgenic (Tg) technologies allowed the engineering of different mouse models of AD recapitulating a range of AD-like features. These Tg models provided excellent opportunities to analyze the bases for the temporal evolution of the disease. Several lines of evidence point to synaptic dysfunction as a cause of AD and that synapse loss is a pathological correlate associated with cognitive decline. Therefore, the phenotypic characterization of these animals has included electrophysiological studies to analyze hippocampal synaptic transmission and long-term potentiation, a widely recognized cellular model for learning and memory. Transgenic mice, along with non-Tg models derived mainly from exogenous application of Aß, have also been useful experimental tools to test the various therapeutic approaches. As a result, numerous pharmacological interventions have been reported to attenuate synaptic dysfunction and improve behavior in the different AD models. To date, however, very few of these findings have resulted in target validation or successful translation into disease-modifying compounds in humans. Here, we will briefly review the synaptic alterations across the different animal models and we will recapitulate the pharmacological strategies aimed at rescuing hippocampal plasticity phenotypes. Finally, we will highlight intrinsic limitations in the use of experimental systems and related challenges in translating preclinical studies into human clinical trials.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Sinapsis/patología , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Humanos , Inmunoterapia , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos
20.
Neuromolecular Med ; 14(4): 262-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22661254

RESUMEN

The insulin receptor (IR) is a protein tyrosine kinase playing a pivotal role in the regulation of peripheral glucose metabolism and energy homoeostasis. IRs are also abundantly distributed in the cerebral cortex and hippocampus, where they regulate synaptic activity required for learning and memory. As the major anabolic hormone in mammals, insulin stimulates protein synthesis partially through the activation of the PI3K/Akt/mTOR pathway, playing fundamental roles in neuronal development, synaptic plasticity and memory. Here, by means of a multidisciplinary approach, we report that long-term synaptic plasticity and recognition memory are impaired in IR ß-subunit heterozygous mice. Since IR expression is diminished in type-2 diabetes as well as in Alzheimer's disease (AD) patients, these data may provide a mechanistic link between insulin resistance, impaired synaptic transmission and cognitive decline in humans with metabolic disorders.


Asunto(s)
Hipocampo/fisiopatología , Discapacidades para el Aprendizaje/genética , Potenciación a Largo Plazo/genética , Trastornos de la Memoria/genética , Proteínas del Tejido Nervioso/deficiencia , Receptor de Insulina/deficiencia , Reconocimiento en Psicología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/psicología , Femenino , Heterocigoto , Humanos , Resistencia a la Insulina , Discapacidades para el Aprendizaje/fisiopatología , Trastornos de la Memoria/fisiopatología , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Densidad Postsináptica/ultraestructura , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptor de Insulina/genética , Receptor de Insulina/fisiología , Transducción de Señal/fisiología , Transmisión Sináptica/genética , Serina-Treonina Quinasas TOR/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...