Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Neurosci ; 21(10): 535-550, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32879508

RESUMEN

Although hormones such as glucocorticoids have been broadly accepted in recent decades as general neuromodulators of memory processes, sex steroid hormones such as the potent oestrogen 17ß-oestradiol have been less well recognized by the scientific community in this capacity. The predominance of females in studies of oestradiol and memory and the general (but erroneous) perception that oestrogens are 'female' hormones have probably prevented oestradiol from being more widely considered as a key memory modulator in both sexes. Indeed, although considerable evidence supports a crucial role for oestradiol in regulating learning and memory in females, a growing body of literature indicates a similar role in males. This Review discusses the mechanisms of oestradiol signalling and provides an overview of the effects of oestradiol on spatial, object recognition, social and fear memories. Although the primary focus is on data collected in females, effects of oestradiol on memory in males will be discussed, as will sex differences in the molecular mechanisms that regulate oestrogenic modulation of memory, which may have important implications for the development of future cognitive therapeutics.


Asunto(s)
Encéfalo/fisiología , Estradiol/fisiología , Aprendizaje/fisiología , Memoria/fisiología , Neuronas/fisiología , Miedo/fisiología , Femenino , Humanos , Masculino , Caracteres Sexuales , Transducción de Señal
2.
Horm Behav ; 165: 105618, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39180889

RESUMEN

Infusion of 17ß-estradiol (E2) into the dorsal hippocampus (DH) of ovariectomized (OVX) mice enhances memory consolidation, an effect that depends on rapid phosphorylation of extracellular signal-regulated kinase (ERK) and Akt. Astrocytic glutamate transporter 1 (GLT-1) modulates neurotransmission via glutamate uptake from the synaptic cleft. However, little is known about the contribution of DH astrocytes, and astrocytic glutamate transport, to the memory-enhancing effects of E2. This study was designed to test whether DH astrocytes contribute to estrogenic modulation of memory consolidation by determining the extent to which DH GLT-1 is necessary for E2 to enhance memory in object recognition and object placement tasks and trigger rapid phosphorylation events in DH astrocytes. OVX female mice were bilaterally cannulated into the DH or the DH and dorsal third ventricle (ICV). Post-training DH infusion of the GLT-1 inhibitor dihydrokainic acid (DHK) dose-dependently impaired memory consolidation in both tasks. Moreover, the memory-enhancing effects of ICV-infused E2 in each task were blocked by DH DHK infusion. E2 increased p42 ERK and Akt phosphorylation in DH astrocytes, and these effects were blocked by DHK. Results suggest the necessity of DH GLT-1 activity for object and spatial memory consolidation, and for E2 to enhance consolidation of these memories and to rapidly activate cell signaling in DH astrocytes. Findings indicate that astrocytic function in the DH of OVX females is necessary for memory formation and is regulated by E2, and suggest an essential role for DH astrocytic GLT-1 activity in the memory-enhancing effects of E2.


Asunto(s)
Astrocitos , Estradiol , Transportador 2 de Aminoácidos Excitadores , Ácido Glutámico , Hipocampo , Ovariectomía , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Femenino , Estradiol/farmacología , Ratones , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ácido Glutámico/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Consolidación de la Memoria/efectos de los fármacos , Consolidación de la Memoria/fisiología , Fosforilación/efectos de los fármacos , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones Endogámicos C57BL , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Ácido Kaínico/análogos & derivados
3.
Horm Behav ; 161: 105516, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38428223

RESUMEN

Studies in ovariectomized (OVX) female rodents suggest that G protein-coupled estrogen receptor (GPER) is a key regulator of memory, yet little is known about its importance to memory in males or the cellular mechanisms underlying its mnemonic effects in either sex. In OVX mice, bilateral infusion of the GPER agonist G-1 into the dorsal hippocampus (DH) enhances object recognition and spatial memory consolidation in a manner dependent on rapid activation of c-Jun N-terminal kinase (JNK) signaling, cofilin phosphorylation, and actin polymerization in the DH. However, the effects of GPER on memory consolidation and DH cell signaling in males are unknown. Thus, the present study first assessed effects of DH infusion of G-1 or the GPER antagonist G-15 on object recognition and spatial memory consolidation in gonadectomized (GDX) male mice. As in OVX mice, immediate post-training bilateral DH infusion of G-1 enhanced, whereas G-15 impaired, memory consolidation in the object recognition and object placement tasks. However, G-1 did not increase levels of phosphorylated JNK (p46, p54) or cofilin in the DH 5, 15, or 30 min after infusion, nor did it affect phosphorylation of ERK (p42, p44), PI3K, or Akt. Levels of phospho-cAMP-responsive element binding protein (CREB) were elevated in the DH 30 min following G-1 infusion, indicating that GPER in males activates a yet unknown signaling mechanism that triggers CREB-mediated gene transcription. Our findings show for the first time that GPER in the DH regulates memory consolidation in males and suggests sex differences in underlying signaling mechanisms.


Asunto(s)
Hipocampo , Consolidación de la Memoria , Quinolinas , Receptores Acoplados a Proteínas G , Transducción de Señal , Animales , Masculino , Consolidación de la Memoria/fisiología , Consolidación de la Memoria/efectos de los fármacos , Femenino , Ratones , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Transducción de Señal/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Ovariectomía , Orquiectomía , Ciclopentanos/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ratones Endogámicos C57BL
4.
Learn Mem ; 29(9): 302-311, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36206392

RESUMEN

The ubiquitin proteasome system (UPS) is a primary mechanism through which proteins are degraded in cells. UPS activity in the dorsal hippocampus (DH) is necessary for multiple types of memory, including object memory, in male rodents. However, sex differences in DH UPS activation after fear conditioning suggest that other forms of learning may also differentially regulate DH UPS activity in males and females. Here, we examined markers of UPS activity in the synaptic and cytoplasmic fractions of DH and medial prefrontal cortex (mPFC) tissue collected 1 h following object training. In males, training increased phosphorylation of proteasomal subunit Rpt6, 20S proteasome activity, and the amount of PSD-95 in the DH synaptic fraction, as well as proteasome activity in the mPFC synaptic fraction. In females, training did not affect measures of UPS or synaptic activity in the DH synaptic fraction or in either mPFC fraction but increased Rpt6 phosphorylation in the DH cytoplasmic fraction. Overall, training-induced UPS activity was greater in males than in females, greater in the DH than in the mPFC, and greater in synaptic fractions than in cytosol. These data suggest that object training drives sex-specific alterations in UPS activity across brain regions and subcellular compartments important for memory.


Asunto(s)
Condicionamiento Clásico , Complejo de la Endopetidasa Proteasomal , Animales , Condicionamiento Clásico/fisiología , Femenino , Hipocampo/fisiología , Masculino , Ratones , Corteza Prefrontal/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Caracteres Sexuales , Ubiquitina/metabolismo
5.
Front Neuroendocrinol ; 62: 100926, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34089761

RESUMEN

The sex steroid hormones (SSHs) play several roles in regulation of various processes in the cardiovascular, immune, muscular and neural systems. SSHs affect prenatal and postnatal development of various brain structures, including regions associated with important physiological, behavioral, cognitive, and emotional functions. This action can be mediated by either intracellular or transmembrane receptors. While the classical mechanisms of SSHs action are relatively well examined, the physiological importance of non-classical mechanism of SSHs action through membrane-associated and transmembrane receptors in the brain remains unclear. The most recent summary describing the role of SSHs in different body systems is lacking. Therefore, the aim of this review is to discuss classical and non-classical signaling pathways of testosterone and estradiol action via their receptors at functional, cellular, tissue level and to describe the effects on various body systems and behavior. Particular emphasis will be on brain regions including the hippocampus, hypothalamus, frontal cortex and cerebellum.


Asunto(s)
Estradiol , Hormonas Esteroides Gonadales , Estrógenos , Femenino , Humanos , Hipotálamo , Embarazo , Testosterona
6.
Horm Behav ; 140: 105124, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35101701

RESUMEN

Anxiety is a prominent and debilitating symptom in Alzheimer's disease (AD) patients. Carriers of APOE4, the greatest genetic risk factor for late-onset AD, may experience increased anxiety relative to carriers of other APOE genotypes. However, whether APOE4 genotype interacts with other AD risk factors to promote anxiety-like behaviors is less clear. Here, we used open field exploration to assess anxiety-like behavior in an EFAD mouse model of AD that expresses five familial AD mutations (5xFAD) and human APOE3 or APOE4. We first examined whether APOE4 genotype exacerbates anxiety-like exploratory behavior in the open field relative to APOE3 genotype in a sex-specific manner among six-month-old male and female E3FAD (APOE3+/+/5xFAD+/-) and E4FAD mice (APOE4+/+/5xFAD+/-). Next, we determined whether circulating ovarian hormone loss influences exploratory behavior in the open field among female E3FAD and E4FADs. APOE4 genotype was associated with decreased time in the center of the open field, particularly among female EFADs. Furthermore, ovariectomy (OVX) decreased time in the center of the open field among female E3FADs to levels similar to intact and OVXed E4FAD females. Our results suggest that APOE4 genotype increased anxiety-like behavior in the open field, and that ovarian hormones may protect against an anxiety-like phenotype in female E3FAD, but not E4FAD mice.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Apolipoproteínas E/genética , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Animales , Apolipoproteína E4/genética , Femenino , Genotipo , Hormonas , Masculino , Ratones , Ratones Transgénicos
7.
Front Neuroendocrinol ; 59: 100860, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32781195

RESUMEN

Cognitive behaviors, such as episodic memory formation, are complex processes involving coordinated activity in multiple brain regions. However, much of the research on hormonal regulation of cognition focuses on manipulation of one region at a time or provides a single snapshot of how a systemic treatment affects multiple brain regions without investigating how these regions might interact to mediate hormone effects. Here, we use estrogenic regulation of episodic memory as an example of how circuit-based approaches may be incorporated into future studies of hormones and cognition. We first review basic episodic memory circuitry, rapid mechanisms by which 17ß-estradiol can alter circuit activity, and current knowledge about 17ß-estradiol's effects on episodic memory. Next, we outline approaches that researchers can employ to consider circuit effects in their estrogen research and provide examples of how these methods have been used to examine hormonal regulation of memory and other behaviors.


Asunto(s)
Estradiol/metabolismo , Hipocampo/fisiología , Memoria Episódica , Corteza Prefrontal/fisiología , Animales , Humanos
8.
Neurobiol Learn Mem ; 185: 107521, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34536525

RESUMEN

Episodic memory is a complex process requiring input from several regions of the brain. Emerging evidence suggests that coordinated activity between the dorsal hippocampus (DH) and medial prefrontal cortex (mPFC) is required for episodic memory consolidation. However, the mechanisms through which the DH and mPFC interact to promote memory consolidation remain poorly understood. A growing body of research suggests that the nucleus reuniens of the thalamus (RE) is one of several structures that facilitate communication between the DH and mPFC during memory and may do so through bidirectional excitatory projections to both regions. Furthermore, recent work from other labs indicates that the RE is necessary for spatial working memory. However, it is not clear to what extent the RE is necessary for memory of object locations. The goal of this study was to determine whether activity in the RE is necessary for spatial memory as measured by the object placement (OP) task in female mice. A kappa-opioid receptor DREADD (KORD) virus was used to inactivate excitatory neurons in the RE pre- or post-training to establish a role for the RE in spatial memory acquisition and consolidation, respectively. RE inactivation prior to, or immediately after, object training blocked OP memory formation relative to chance and to control mice. Moreover, expression of the immediate early gene EGR-1 was reduced in the RE 1 hour after an object training trial, supporting the conclusion that reduced neuronal activity in the RE impairs the formation of object location memories. In summary, the findings of this study support a key role for the RE in spatial memory acquisition and consolidation.


Asunto(s)
Núcleos Talámicos de la Línea Media/fisiología , Memoria Espacial/fisiología , Animales , Diterpenos de Tipo Clerodano/farmacología , Hipocampo/fisiología , Consolidación de la Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Núcleos Talámicos de la Línea Media/anatomía & histología , Núcleos Talámicos de la Línea Media/efectos de los fármacos , Corteza Prefrontal/fisiología
9.
Horm Behav ; 130: 104948, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33571507

RESUMEN

Development of estrogen therapies targeting the ß (ERß) but not α (ERα) estrogen receptor is critically needed for the treatment of negative menopausal symptoms, as ERα activation increases health risks like cancer. Here, we determined the effects of long-term oral treatment with EGX358, a novel highly selective ERß agonist, on memory, vasodilation, and affect in young ovariectomized mice. Mice were orally gavaged daily for 9 weeks with vehicle, 17ß-estradiol (E2), the ERß agonist diarylpropionitrile (DPN), or EGX358 at doses that enhance memory when delivered acutely. Tail skin temperature was recorded as a proxy for vasodilation following injection of vehicle or senktide, a tachykinin receptor 3 agonist used to model hot flashes. Anxiety-like behavior was assessed in the open field (OF) and elevated plus maze (EPM), and depression-like behavior was measured in the tail suspension (TST) and forced swim tests (FST). Finally, memory was assessed in object recognition (OR) and object placement (OP) tasks. E2, DPN, and EGX358 reduced senktide-mediated increases in tail skin temperature compared to vehicle. All three treatments also enhanced memory in the OR and OP tasks, whereas vehicle did not. Although E2 increased time spent in the center of the OF, no other treatment effects were observed in the OF, EPM, TST, or FST. These data suggest that long-term ERß activation can reduce hot flash-like symptoms and enhance spatial and object recognition memories in ovariectomized mice. Thus, the highly selective ERß agonist EGX358 may be a promising avenue for reducing menopause-related hot flashes and memory dysfunction.


Asunto(s)
Receptor beta de Estrógeno , Preparaciones Farmacéuticas , Administración Oral , Animales , Estradiol/farmacología , Receptor alfa de Estrógeno , Femenino , Humanos , Ratones , Nitrilos/farmacología , Ovariectomía , Vasodilatación
10.
J Neurosci ; 39(48): 9598-9610, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31628182

RESUMEN

Activation of the membrane estrogen receptor G-protein-coupled estrogen receptor (GPER) in ovariectomized mice via the GPER agonist G-1 mimics the beneficial effects of 17ß-estradiol (E2) on hippocampal CA1 spine density and memory consolidation, yet the cell-signaling mechanisms mediating these effects remain unclear. The present study examined the role of actin polymerization and c-Jun N-terminal kinase (JNK) phosphorylation in mediating effects of dorsal hippocampally infused G-1 on CA1 dendritic spine density and consolidation of object recognition and spatial memories in ovariectomized mice. We first showed that object learning increased apical CA1 spine density in the dorsal hippocampus (DH) within 40 min. We then found that DH infusion of G-1 increased both CA1 spine density and phosphorylation of the actin polymerization regulator cofilin, suggesting that activation of GPER may increase spine morphogenesis through actin polymerization. As with memory consolidation in our previous work (Kim et al., 2016), effects of G-1 on CA1 spine density and cofilin phosphorylation depended on JNK phosphorylation in the DH. Also consistent with our previous findings, E2-induced cofilin phosphorylation was not dependent on GPER activation. Finally, we found that infusion of the actin polymerization inhibitor, latrunculin A, into the DH prevented G-1 from increasing apical CA1 spine density and enhancing both object recognition and spatial memory consolidation. Collectively, these data demonstrate that GPER-mediated hippocampal spinogenesis and memory consolidation depend on JNK and cofilin signaling, supporting a critical role for actin polymerization in the GPER-induced regulation of hippocampal function in female mice.SIGNIFICANCE STATEMENT Emerging evidence suggests that G-protein-coupled estrogen receptor (GPER) activation mimics effects of 17ß-estradiol on hippocampal memory consolidation. Unlike canonical estrogen receptors, GPER activation is associated with reduced cancer cell proliferation; thus, understanding the molecular mechanisms through which GPER regulates hippocampal function may provide new avenues for the development of drugs that provide the cognitive benefits of estrogens without harmful side effects. Here, we demonstrate that GPER increases CA1 dendritic spine density and hippocampal memory consolidation in a manner dependent on actin polymerization and c-Jun N-terminal kinase phosphorylation. These findings provide novel insights into the role of GPER in mediating hippocampal morphology and memory consolidation, and may suggest first steps toward new therapeutics that more safely and effectively reduce memory decline in menopausal women.


Asunto(s)
Actinas/metabolismo , Región CA1 Hipocampal/metabolismo , Espinas Dendríticas/metabolismo , Consolidación de la Memoria/fisiología , Polimerizacion , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Actinas/análisis , Animales , Región CA1 Hipocampal/química , Espinas Dendríticas/química , Femenino , Ratones , Ratones Endogámicos C57BL , Receptores de Estrógenos/análisis , Receptores Acoplados a Proteínas G/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA