Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cereb Cortex ; 30(4): 2627-2641, 2020 04 14.
Article in English | MEDLINE | ID: mdl-31800024

ABSTRACT

Numerous studies provide increasing evidence, which supports the ideas that every cell in the brain of males may differ from those in females due to differences in sex chromosome complement as well as in response to hormonal effects. In this study, we address the question as to whether actions of neurosteroids, thus steroids, which are synthesized and function within the brain, contribute to sex-specific hippocampal synaptic plasticity. We have previously shown that predominantly in the female hippocampus, does inhibition of the conversion of testosterone to estradiol affect synaptic transmission. In this study, we show that testosterone and its metabolite dihydrotestosterone are essential for hippocampal synaptic transmission specifically in males. This also holds true for the density of mushroom spines and of spine synapses. We obtained similar sex-dependent results using primary hippocampal cultures of male and female animals. Since these cultures originated from perinatal animals, our findings argue for sex-dependent differentiation of hippocampal neurons regarding their responsiveness to sex neurosteroids up to birth, which persist during adulthood. Hence, our in vitro findings may point to a developmental effect either directly induced by sex chromosomes or indirectly by fetal testosterone secretion during the perinatal critical period, when developmental sexual priming takes place.


Subject(s)
Hippocampus/metabolism , Neuronal Plasticity/physiology , Neurosteroids/metabolism , Sex Characteristics , Synapses/metabolism , Animals , Cells, Cultured , Female , Hippocampus/ultrastructure , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Rats , Rats, Wistar , Synapses/ultrastructure
2.
Sci Rep ; 8(1): 8722, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880879

ABSTRACT

Reelin plays an important role in cerebral cortex development and synaptogenesis. In the hippocampus, the neurosteroid estrogen affects reelin expression. In this study we tested a potential crosstalk between estradiol and reelin, thus the possibility of a reelin-induced activation of the estradiol synthesizing enzyme aromatase. As a model system, we used ovaries, which express reelin and are a major source of estradiol. We found that in wild-type mice, reelin and aromatase are expressed in granulosa cells of growing follicles. The expression of reelin varies with the estrus cycle and is highest shortly before ovulation, when estradiol serum levels are at their maximum. In ovaries of reelin-deficient reeler mice, aromatase mRNA and protein are significantly reduced, as evidenced by real-time PCR, western blot analysis, and quantitative immunohistochemistry in granulosa cells of preovulatory follicles. In line with reduced estradiol synthesis, ovarian estrus cycle length is prolonged in reeler mice. Most importantly, treating cultured granulosa cells with recombinant reelin results in significant upregulation of aromatase mRNA and protein and increased secretion of estradiol into the supernatant. Our data provide evidence of a local increase of aromatase expression by reelin. Regarding reproduction, this crosstalk may contribute to follicular stability and counteract luteinization in ovaries.


Subject(s)
Aromatase/biosynthesis , Cell Adhesion Molecules, Neuronal/biosynthesis , Estrous Cycle/physiology , Extracellular Matrix Proteins/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Granulosa Cells/metabolism , Luteinization/physiology , Nerve Tissue Proteins/biosynthesis , Serine Endopeptidases/biosynthesis , Animals , Female , Granulosa Cells/cytology , Mice , Rats, Wistar , Reelin Protein
3.
Brain Pathol ; 28(1): 14-27, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27880990

ABSTRACT

Nowadays, amyotrophic lateral sclerosis (ALS) is considered as a multisystem disorder, characterized by a primary degeneration of motor neurons as well as neuropathological changes in non-motor regions. Neurodegeneration in subcortical areas, such as the thalamus, are believed to contribute to cognitive and behavioral abnormalities in ALS patients. In the present study, we investigated neurodegenerative changes including neuronal loss and glia pathology in the anterodorsal thalamic nucleus (AD) of SOD1(G93A) mice, a widely used animal model for ALS. We detected massive dendrite swelling and neuronal loss in SOD1(G93A) animals, which was accompanied by a mild gliosis. Furthermore, misfolded SOD1 protein and autophagy markers were accumulating in the AD. Since innate immunity and activation inflammasomes seem to play a crucial role in ALS, we examined protein expression of Nod-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase-1 recruitment domain (ASC) and the cytokine interleukin 1 beta (IL1ß) in AD glial cells and neurons. NLRP3 and ASC were significantly up-regulated in the AD of SOD1(G93A) mice. Finally, co-localization studies revealed expression of NLRP3, ASC and IL1ß in neurons. Our study yielded two main findings: (i) neurodegenerative changes already occur at an early symptomatic stage in the AD and (ii) increased inflammasome expression may contribute to neuronal cell death. In conclusion, neurodegeneration in the anterior thalamus may critically account for cognitive changes in ALS pathology.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Anterior Thalamic Nuclei/pathology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nerve Degeneration/pathology , Neurons/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Anterior Thalamic Nuclei/physiopathology , CARD Signaling Adaptor Proteins/metabolism , Cell Death/physiology , Disease Models, Animal , Disease Progression , Female , Humans , Interleukin-1beta/metabolism , Male , Mice, Transgenic , Nerve Degeneration/physiopathology , Neuroglia/pathology , Neuroglia/physiology , Neurons/physiology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
4.
Learn Mem ; 24(12): 650-659, 2017 12.
Article in English | MEDLINE | ID: mdl-29142062

ABSTRACT

The serine protease inhibitor neuroserpin regulates the activity of tissue-type plasminogen activator (tPA) in the nervous system. Neuroserpin expression is particularly prominent at late stages of neuronal development in most regions of the central nervous system (CNS), whereas it is restricted to regions related to learning and memory in the adult brain. The physiological expression pattern of neuroserpin, its high degree of colocalization with tPA within the CNS, together with its dysregulation in neuropsychiatric disorders, suggest a role in formation and refinement of synapses. In fact, studies in cell culture and mice point to a role for neuroserpin in dendritic branching, spine morphology, and modulation of behavior. In this study, we investigated the physiological role of neuroserpin in the regulation of synaptic density, synaptic plasticity, and behavior in neuroserpin-deficient mice. In the absence of neuroserpin, mice show a significant decrease in spine-synapse density in the CA1 region of the hippocampus, while expression of the key postsynaptic scaffold protein PSD-95 is increased in this region. Neuroserpin-deficient mice show decreased synaptic potentiation, as indicated by reduced long-term potentiation (LTP), whereas presynaptic paired-pulse facilitation (PPF) is unaffected. Consistent with altered synaptic plasticity, neuroserpin-deficient mice exhibit cognitive and sociability deficits in behavioral assays. However, although synaptic dysfunction is implicated in neuropsychiatric disorders, we do not detect alterations in expression of neuroserpin in fusiform gyrus of autism patients or in dorsolateral prefrontal cortex of schizophrenia patients. Our results identify neuroserpin as a modulator of synaptic plasticity, and point to a role for neuroserpin in learning and memory.


Subject(s)
Gene Expression Regulation/genetics , Neuronal Plasticity/genetics , Neuropeptides/deficiency , Serine Proteinase Inhibitors/metabolism , Serpins/deficiency , Social Behavior , Synapses/genetics , Adolescent , Adult , Animals , Autistic Disorder/genetics , Autistic Disorder/pathology , Autistic Disorder/psychology , Child , Exploratory Behavior/physiology , Hippocampus/physiology , Hippocampus/ultrastructure , Humans , Long-Term Potentiation/genetics , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neuropeptides/genetics , Serpins/genetics , Synapses/physiology , Synapses/ultrastructure , Synaptosomal-Associated Protein 25/metabolism , Young Adult , Neuroserpin
5.
J Neurosci ; 37(6): 1532-1545, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28028198

ABSTRACT

The basolateral amygdala (BLA) integrates sensory input from cortical and subcortical regions, a function that requires marked synaptic plasticity. Here we provide evidence that cytochrome P450 aromatase (AROM), the enzyme converting testosterone to 17ß-estradiol (E2), contributes to the regulation of this plasticity in a sex-specific manner. We show that AROM is expressed in the BLA, particularly in the basolateral nucleus (BL), in male and female rodents. Systemic administration of the AROM inhibitor letrozole reduced spine synapse density in the BL of adult female mice but not in the BL of male mice. Similarly, in organotypic corticoamygdalar slice cultures from immature rats, treatment with letrozole significantly reduced spine synapses in the BL only in cultures derived from females. In addition, letrozole sex-specifically altered synaptic properties in the BL: in acute slices from juvenile (prepubertal) female rats, wash-in of letrozole virtually abolished long-term potentiation (LTP), whereas it did not prevent the generation of LTP in the slices from males. Together, these data indicate that neuron-derived E2 modulates synaptic plasticity in rodent BLA sex-dependently. As protein expression levels of AROM, estrogen and androgen receptors did not differ between males and females and were not sex-specifically altered by letrozole, the findings suggest sex-specific mechanisms of E2 signaling.SIGNIFICANCE STATEMENT The basolateral amygdala (BLA) is a key structure of the fear circuit. This research reveals a sexually dimorphic regulation of synaptic plasticity in the BLA involving neuronal aromatase, which produces the neurosteroid 17ß-estradiol (E2). As male and female neurons in rodent BLA responded differently to aromatase inhibition both in vivo and in vitro, our findings suggest that E2 signaling in BLA neurons is regulated sex-dependently, presumably via mechanisms that have been established during sexual determination. These findings could be relevant for the understanding of sex differences in mood disorders and of the side effects of cytochrome P450 aromatase inhibitors, which are frequently used for breast cancer therapy.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/physiology , Basolateral Nuclear Complex/physiology , Neuronal Plasticity/physiology , Sex Characteristics , Animals , Basolateral Nuclear Complex/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Letrozole , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Nitriles/pharmacology , Organ Culture Techniques , Rats , Triazoles/pharmacology
6.
J Neurochem ; 140(1): 126-139, 2017 01.
Article in English | MEDLINE | ID: mdl-27861893

ABSTRACT

Locally synthesized estradiol plays an important role in synaptic plasticity in the hippocampus. We have previously shown that in hippocampal neurons, activity of the enzyme aromatase, which converts testosterone into estradiol, is reduced via Ca2+ -dependent phosphorylation. Synaptopodin is a highly estrogen responsive protein, and it has been shown that it is an important regulator of synaptic plasticity, mediated by its close association with internal calcium stores. In this study, we show that the expression of synaptopodin is stronger in the hippocampus of female animals than in that of male animals. Phosphorylation of aromatase, using letrozole, however, down-regulates synaptopodin immunohistochemistry in the hippocampus of both male and females. Similarly, in aromatase knock-out mice synaptopodin expression in the hippocampus is reduced sex independently. Using primary-dissociated hippocampal neurons, we found that evoked release of Ca2+ from internal stores down-regulates aromatase activity, which is paralleled by reduced expression of synaptopodin. Opposite effects were achieved after inhibition of the release. Calcium-dependent regulation of synaptopodin expression was abolished when the control of aromatase activity by the Ca2+ transients was disrupted. Our data suggest that the regulation of aromatase activity by Ca2+ transients in neurons contributes to synaptic plasticity in the hippocampus of male and female animals as an on-site regulatory mechanism.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/metabolism , Microfilament Proteins/physiology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Hippocampus/drug effects , Hippocampus/metabolism , Letrozole , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitriles/pharmacology , Rats , Rats, Wistar , Triazoles/pharmacology
7.
Prog Histochem Cytochem ; 48(2): 49-64, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23906992

ABSTRACT

Estradiol synthesis in the ovaries is regulated via feedback mechanisms mediated by gonadotrophin-releasing hormone (GnRH) and gonadotrophins, secreted by the hypothalamus and the pituitary, respectively. Estradiol synthesis also takes place in the hippocampus. In hippocampal slice cultures of female animals, GnRH regulates estradiol synthesis dose-dependently. Hence, both hippocampal and ovarian estradiol synthesis are synchronized by GnRH. Hippocampus-derived estradiol is essential to synapse stability and maintenance because it stabilizes the spine cytoskeleton of hippocampal neurons. Inhibition of hippocampal estradiol synthesis in mice, however, results in loss of spines and spine synapses in females, but not in males. Stereotaxic application of GnRH to the hippocampus of female rats confirms the regulatory role of GnRH on estradiol synthesis and synapse density in the female hippocampus in vivo. This regulatory role of GnRH necessarily results in estrus cyclicity of spine density in the hippocampus of females.


Subject(s)
Estradiol/biosynthesis , Gonadotropin-Releasing Hormone/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Animals , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dose-Response Relationship, Drug , Female , Hippocampus/cytology , Immunohistochemistry , Male , Mice , Neurons/drug effects , Neurons/metabolism , Rats , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Synapses/drug effects , Synapses/metabolism , Tissue Culture Techniques
8.
J Neurosci ; 32(24): 8116-26, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22699893

ABSTRACT

Inhibitors of aromatase, the final enzyme of estradiol synthesis, are suspected of inducing memory deficits in women. In previous experiments, we found hippocampal spine synapse loss in female mice that had been treated with letrozole, a potent aromatase inhibitor. In this study, we therefore focused on the effects of letrozole on long-term potentiation (LTP), which is an electrophysiological parameter of memory and is known to induce spines, and on phosphorylation of cofilin, which stabilizes the spine cytoskeleton and is required for LTP in mice. In acute slices of letrozole-treated female mice with reduced estradiol serum concentrations, impairment of LTP started as early as after 6 h of treatment and progressed further, together with dephosphorylation of cofilin in the same slices. Theta-burst stimulation failed to induce LTP after 1 week of treatment. Impairment of LTP was followed by spine and spine synapse loss. The effects were confirmed in vitro by using hippocampal slice cultures of female mice. The sequence of effects in response to letrozole were similar in ovariectomized female and male mice, with, however, differences as to the degree of downregulation. Our data strongly suggest that impairment of LTP, followed by loss of mushroom spines and spine synapses in females, may have implications for memory deficits in women treated with letrozole.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/physiology , Long-Term Potentiation/physiology , Nitriles/pharmacology , Sex Characteristics , Triazoles/pharmacology , Animals , Cells, Cultured , Cofilin 1/metabolism , Dendritic Spines/drug effects , Dendritic Spines/ultrastructure , Estradiol/blood , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Letrozole , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Synapses/drug effects , Synapses/physiology , Synapses/ultrastructure
9.
J Steroid Biochem Mol Biol ; 131(1-2): 24-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22138012

ABSTRACT

Hippocampal neurons are capable of synthesizing estradiol de novo. Estradiol synthesis can be suppressed by aromatase inhibitors and by knock-down of steroid acute regulatory protein (StAR), whereas elevated levels of substrates of steroidogenesis enhance estradiol synthesis. In rat hippocampal cultures, the expression of estrogen receptors (ERs) and synaptic proteins, as well as synapse density, correlated positively with aromatase activity, regardless of whether the cultures originated from males or females. All effects induced by the inhibition of aromatase activity were rescued by application of estradiol to the cultures. In vivo, however, systemic application of letrozole, an aromatase inhibitor, induced synapse loss in female rats, but not in males. Furthermore, in the female hippocampus, density of spines and spine synapses varied with the estrus cycle. In addressing this in vivo-in vitro discrepancy, we found that gonadotropin-releasing hormone (GnRH) regulated estradiol synthesis via an aromatase-mediated mechanism and consistently regulated spine synapse density and the expression of synaptic proteins. Along these lines, GnRH receptor density was higher in the hippocampus than in the cortex and hypothalamus, and estrus cyclicity of spinogenesis was found in the hippocampus, but not in the cortex. Since GnRH receptor expression also varies with the estrus cycle, the sexual dimorphism in estrogen-regulated spine synapse density in the hippocampus very likely results from differences in the GnRH responsiveness of the male and the female hippocampus. This article is part of a Special Issue entitled 'Neurosteroids'.


Subject(s)
Estradiol/physiology , Hippocampus/drug effects , Hippocampus/physiology , Synapses/drug effects , Synapses/physiology , Animals , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , Cells, Cultured , Cerebral Cortex/metabolism , Dendritic Spines/drug effects , Dendritic Spines/physiology , Estradiol/biosynthesis , Estrus/physiology , Female , Hypothalamus/metabolism , Letrozole , Male , Nitriles/pharmacology , Rats , Receptors, Estrogen/biosynthesis , Sex Factors , Triazoles/pharmacology
10.
J Mol Neurosci ; 45(3): 432-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21822709

ABSTRACT

The accumulation of proteins such as Tau is a hallmark of several neurodegenerative diseases, e.g., frontotemporal dementia (FTD). So far, many mouse models of tauopathies have been generated by the use of mutated or truncated human Tau isoforms in order to enhance the amyloidogenic character of Tau and to mimic pathological processes similar to those in FTD patients. Our inducible mice express the repeat domain of human Tau (Tau(RD)) carrying the FTDP-17 mutation ΔK280 in a "pro-aggregant" and an "anti-aggregant" version. Based on the enhanced tendency of Tau to aggregate, only the "pro-aggregant" Tau(RD) mice develop Tau pathology (hyperphosphorylation, coassembly of human and mouse Tau, synaptic loss, and neuronal degeneration). We have now carried out behavioral and electrophysiological analyses showing that only the pro-aggregant Tau(RD) mice have impaired learning/memory and a distinct loss of LTP. Remarkably, after suppressing the pro-aggregant human Tau(RD), memory and LTP recover, while neuronal loss persists. Aggregates persist as well but change their composition from mixed human/mouse to mouse Tau only. The rescue of cognition and synaptic plasticity is explained by a partial recovery of spine synapses in the hippocampus. These results indicate a tight relationship between the amyloidogenic character of Tau and brain malfunction, and suggest that the cognitive impairment is caused by toxic human Tau(RD) species rather than by mouse Tau aggregates.


Subject(s)
Cognition Disorders/physiopathology , Disease Models, Animal , Mice, Transgenic , Tauopathies/physiopathology , tau Proteins/metabolism , Animals , Cognition Disorders/pathology , Humans , Learning/physiology , Long-Term Potentiation/physiology , Memory/physiology , Mice , Mutation , Tauopathies/genetics , Tauopathies/pathology , tau Proteins/chemistry , tau Proteins/genetics
11.
J Neurosci ; 31(7): 2511-25, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21325519

ABSTRACT

This report describes the behavioral and electrophysiological analysis of regulatable transgenic mice expressing mutant repeat domains of human Tau (Tau(RD)). Mice were generated to express Tau(RD) in two forms, differing in their propensity for ß-structure and thus in their tendency for aggregation ("pro-aggregant" or "anti-aggregant") (Mocanu et al., 2008). Only pro-aggregant mice show pronounced changes typical for Tau pathology in Alzheimer's disease (aggregation, missorting, hyperphosphorylation, synaptic and neuronal loss), indicating that the ß-propensity and hence the ability to aggregate is a key factor in the disease. We now tested the mice with regard to neuromotor parameters, behavior, learning and memory, and synaptic plasticity and correlated this with histological and biochemical parameters in different stages of switching Tau(RD) on or off. The mice are normal in neuromotor tests. However, pro-aggregant Tau(RD) mice are strongly impaired in memory and show pronounced loss of long-term potentiation (LTP), suggesting that Tau aggregation specifically perturbs these brain functions. Remarkably, when the expression of human pro-aggregant Tau(RD) is switched on for ∼ 10 months and off for ∼ 4 months, memory and LTP recover, whereas aggregates decrease moderately and change their composition from mixed human plus mouse Tau to mouse Tau only. Neuronal loss persists, but synapses are partially rescued. This argues that continuous presence of amyloidogenic pro-aggregant Tau(RD) constitutes the main toxic insult for memory and LTP, rather than the aggregates as such.


Subject(s)
Long-Term Potentiation/genetics , Maze Learning/physiology , Memory/physiology , Mutation/genetics , Tauopathies , tau Proteins/genetics , Animals , Disease Models, Animal , Doxycycline/pharmacology , Hand Strength/physiology , Hippocampus/pathology , Hippocampus/ultrastructure , Humans , In Vitro Techniques , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropsychological Tests , Protein Structure, Tertiary , Space Perception/physiology , Synapses/pathology , Synapses/ultrastructure , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/physiopathology , Time Factors , tau Proteins/metabolism
12.
Cereb Cortex ; 20(12): 2985-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20421250

ABSTRACT

Studies on the role of 17ß-estradiol (E2) in the hippocampus have mainly focused on CA1 and CA3 regions, whereas in dentate gyrus (DG), its role is largely unknown. Here, we examined potential functions of E2 in DG, particularly during development. Immunohistochemistry and in situ hybridization revealed abundance of estrogen receptor (ER)α, but not ERß, expression in DG. Similar to CA1, analysis of synapse densities revealed a reduction in spine synapse number in DG molecular layer of immature rats and adult mice after inhibition of estradiol synthesis using letrozole. Interestingly, strong expression of ERα was found in Cajal-Retzius (CR) cells, which regulate neuronal migration and synaptogenesis via the extracellular matrix protein reelin. Immunoreactivity of aromatase, the final enzyme of estradiol synthesis, was strongest in mature granule cells. In hippocampal slice cultures, exogenous application of E2 caused an increase in reelin expression in CR cells, which was abolished after blockade of ERs using ICI182,780. Vice versa, inhibition of aromatase activity by letrozole resulted in reduced reelin expression, suggesting that E2 deriving from hippocampal sources contributes to the regulation of reelin as well as to the maintenance of spine synapses in DG. E2 further regulated Notch1, a signaling protein involved in neuronal differentiation.


Subject(s)
Cell Adhesion Molecules, Neuronal/biosynthesis , Dentate Gyrus/metabolism , Estradiol/metabolism , Extracellular Matrix Proteins/biosynthesis , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/biosynthesis , Neurogenesis/physiology , Serine Endopeptidases/biosynthesis , Animals , Blotting, Western , Dentate Gyrus/growth & development , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Hippocampus/growth & development , Hippocampus/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Organ Culture Techniques , Rats , Rats, Wistar , Reelin Protein , Synapses
13.
Endocrinology ; 151(3): 1153-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20097718

ABSTRACT

Recently, inhibition of estrogen synthesis by aromatase inhibitors has become a favored therapy for breast cancer in postmenopausal women. Estrogen is, however, important for synapse formation in the hippocampus. Inhibition of aromatase induces spine synapse loss in organotypic hippocampal slice cultures. We therefore studied the effect of systemic treatment with the potent aromatase inhibitor letrozole on spine synapse formation and synaptic proteins in the hippocampi of female mice for periods of 7 d and 4 wk. In cyclic, letrozole-treated females and in ovariectomized, letrozole-treated females, the number of spine synapses was significantly reduced in the hippocampus but not in the prefrontal or cerebellar cortex. Consequently, the expression of the N-methyl-D-aspartate receptor NR1 was significantly down-regulated after treatment with letrozole. In cyclic animals the expression of the synaptic proteins synaptophysin and spinophilin was down-regulated in response to letrozole. In ovariectomized animals, however, protein expression was down-regulated after 7 d of treatment, whereas the expression was up-regulated after 4 wk of treatment. Our results indicate that systemic inhibition of aromatase in mice affects structural synaptic plasticity in the hippocampus. This may contribute to cognitive deficits in postmenopausal women treated with aromatase inhibitors.


Subject(s)
Aromatase Inhibitors/adverse effects , Hippocampus/drug effects , Nitriles/adverse effects , Synapses/drug effects , Triazoles/adverse effects , Animals , Aromatase Inhibitors/administration & dosage , Down-Regulation , Estrous Cycle , Female , Hippocampus/ultrastructure , Letrozole , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nitriles/administration & dosage , Ovariectomy , Synaptophysin/metabolism , Triazoles/administration & dosage
14.
Psychoneuroendocrinology ; 34 Suppl 1: S123-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19781860

ABSTRACT

Estrogen-induced synaptic plasticity was frequently shown by an increase of spines at apical dendrites of CA1 pyramidal neurons after systemic application of estradiol to ovariectomized rats. Surprisingly, exogenous application of estradiol to hippocampal cultures had no effect on spines and on spine synapses, although quantitative immunohistochemistry revealed an upregulation of spinophilin and of synaptophysin, in these cultures. The role of synaptophysin as a presynaptic marker and of spinophilin as a postsynaptic marker, appears questionable from these discrepancies. In contrast, synaptopodin, a marker protein of "mature" mushroom-shaped spines, was downregulated after treatment of hippocampal cultures with estradiol. Synaptopodin is strongly associated to the spine apparatus, a spine-specific cell organelle, which is present in 80% of all mushroom-shaped spines. Consistently, we found a reduction in the number of spines, containing a spine apparatus in response to estradiol, suggesting that the presence of a spine apparatus in many but not all spines is very likely a result of their dynamic character. In summary, synaptic proteins appear to be regulated by estradiol, independent of its function on spine and spine synapse formation.


Subject(s)
Estradiol/pharmacology , Estradiol/physiology , Hippocampus/drug effects , Synapses/metabolism , Animals , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Down-Regulation/drug effects , Hippocampus/metabolism , Mice , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Synapses/ultrastructure , Synaptophysin/metabolism , Up-Regulation/drug effects
15.
J Neural Transm (Vienna) ; 116(11): 1417-25, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19730783

ABSTRACT

Hippocampal spine density varies with the estrus cycle. The cyclic change in estradiol levels in serum was hypothesized to underlie this phenomenon, since treatment of ovariectomized animals with estradiol induced an increase in spine density in hippocampal dendrites of rats, as compared to ovariectomized controls. In contrast, application of estradiol to hippocampal slice cultures did not promote spinogenesis. In addressing this discrepancy, we found that hippocampal neurons themselves are capable of synthesizing estradiol de novo. Estradiol synthesis can be suppressed by aromatase inhibitors and by knock-down of Steroid Acute Regulatory Protein (StAR) and enhanced by substrates of steroidogenesis. Expression of estrogen receptors (ERs) and synaptic proteins, synaptogenesis, and long-term potentiation (LTP) correlated positively with aromatase activity in hippocampal cultures without any difference between genders. All effects due to inhibition of aromatase activity were rescued by application of estradiol to the cultures. Most importantly, gonadotropin-releasing hormone (GnRH) increased estradiol synthesis dose-dependently via an aromatase-mediated mechanism and consistently increased spine synapse density and spinophilin expression. As a consequence, our data suggest that cyclic fluctuations in spine synapse density result from pulsative release of GnRH from the hypothalamus and its effect on hippocampal estradiol synthesis, rather than from varying levels of serum estradiol. This hypothesis is further supported by higher GnRH receptor (GnRH-R) density in the hippocampus than in the cortex and hypothalamus and the specificity of estrus cyclicity of spinogenesis in the hippocampus, as compared to the cortex.


Subject(s)
Cerebral Cortex/metabolism , Dendritic Spines/metabolism , Estrogens/biosynthesis , Estrus/physiology , Hippocampus/metabolism , Neuronal Plasticity/physiology , Animals , Aromatase/drug effects , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , Cerebral Cortex/cytology , Dendritic Spines/ultrastructure , Female , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Hippocampus/cytology , Hypothalamus/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Phosphoproteins/metabolism , Rats , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism
16.
Hippocampus ; 19(8): 692-705, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19156851

ABSTRACT

Cholesterol of glial origin promotes synaptogenesis (Mauch et al., (2001) Science 294:1354-1357). Because in the hippocampus local estradiol synthesis is essential for synaptogenesis, we addressed the question of whether cholesterol-promoted synapse formation results from the function of cholesterol as a precursor of estradiol synthesis in this brain area. To this end, we treated hippocampal cultures with cholesterol, estradiol, or with letrozole, a potent aromatase inhibitor. Cholesterol increased neuronal estradiol release into the medium, the number of spine synapses in hippocampal slice cultures, and immunoreactivity of synaptic proteins in dispersed cultures. Simultaneous application of cholesterol and letrozole or blockade of estrogen receptors by ICI 182 780 abolished cholesterol-induced synapse formation. As a further approach, we inhibited the access of cholesterol to the first enzyme of steroidogenesis by knock-down of steroidogenic acute regulatory protein, the rate-limiting step in steroidogenesis. A rescue of reduced synaptic protein expression in transfected cells was achieved by estradiol but not by cholesterol. Our data indicate that in the hippocampus cholesterol-promoted synapse formation requires the conversion of cholesterol to estradiol.


Subject(s)
Cholesterol/metabolism , Estradiol/metabolism , Hippocampus/physiology , Neurons/physiology , Synapses/physiology , Animals , Animals, Newborn , Aromatase Inhibitors/pharmacology , Cells, Cultured , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Female , Fulvestrant , Hippocampus/drug effects , Hippocampus/ultrastructure , In Vitro Techniques , Letrozole , Male , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/ultrastructure , Nitriles/pharmacology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Rats , Rats, Wistar , Synapses/drug effects , Synapses/ultrastructure , Synaptophysin/metabolism , Triazoles/pharmacology
17.
J Neurosci ; 28(3): 737-48, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18199773

ABSTRACT

We describe two new transgenic mouse lines for studying pathological changes of Tau protein related to Alzheimer's disease. They are based on the regulatable expression of the four-repeat domain of human Tau carrying the FTDP17 (frontotemporal dementia and parkinsonism linked to chromosome 17) mutation deltaK280 (Tau(RD)/deltaK280), or the deltaK280 plus two proline mutations in the hexapeptide motifs (Tau(RD)/deltaK280/I277P/I308P). The deltaK280 mutation accelerates aggregation ("proaggregation mutant"), whereas the proline mutations inhibit Tau aggregation in vitro and in cell models ("antiaggregation mutant"). The inducible transgene expression was driven by the forebrain-specific CaMKIIalpha (calcium/calmodulin-dependent protein kinase IIalpha) promoter. The proaggregation mutant leads to Tau aggregates and tangles as early as 2-3 months after gene expression, even at low expression (70% of endogenous mouse Tau). The antiaggregation mutant does not aggregate even after 22 months of gene expression. Both mutants show missorting of Tau in the somatodendritic compartment and hyperphosphorylation in the repeat domain [KXGS motifs, targets of the kinase MARK (microtubule affinity regulating kinase)]. This indicates that these changes are related to Tau expression rather than aggregation. The proaggregation mutant causes astrogliosis, loss of synapses and neurons from 5 months of gene expression onward, arguing that Tau toxicity is related to aggregation. Remarkably, the human proaggregation mutant Tau(RD) coaggregates with mouse Tau, coupled with missorting and hyperphosphorylation at multiple sites. When expression of proaggregation Tau(RD) is switched off, soluble and aggregated exogenous Tau(RD) disappears within 1.5 months. However, tangles of mouse Tau, hyperphosphorylation, and missorting remain, suggesting an extended lifetime of aggregated wild-type Tau once a pathological conformation and aggregation is induced by a proaggregation Tau species.


Subject(s)
Neurons/pathology , Synapses/pathology , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/chemistry , Age Factors , Animals , Cell Death/physiology , Detergents/pharmacology , Disease Models, Animal , Gene Expression/physiology , Humans , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Mutation/physiology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/ultrastructure , Phosphorylation , Protein Structure, Tertiary , Sarcosine/analogs & derivatives , Sarcosine/pharmacology , Silver Staining/methods , Synapses/metabolism , Synapses/ultrastructure , tau Proteins/drug effects , tau Proteins/genetics
18.
J Cell Biol ; 180(2): 417-26, 2008 Jan 28.
Article in English | MEDLINE | ID: mdl-18227283

ABSTRACT

Spine density in the hippocampus changes during the estrus cycle and is dependent on the activity of local aromatase, the final enzyme in estrogen synthesis. In view of the abundant gonadotropin-releasing hormone receptor (GnRH-R) messenger RNA expression in the hippocampus and the direct effect of GnRH on estradiol (E2) synthesis in gonadal cells, we asked whether GnRH serves as a regulator of hippocampal E2 synthesis. In hippocampal cultures, E2 synthesis, spine synapse density, and immunoreactivity of spinophilin, a reliable spine marker, are consistently up-regulated in a dose-dependent manner at low doses of GnRH but decrease at higher doses. GnRH is ineffective in the presence of GnRH antagonists or aromatase inhibitors. Conversely, GnRH-R expression increases after inhibition of hippocampal aromatase. As we found estrus cyclicity of spine density in the hippocampus but not in the neocortex and GnRH-R expression to be fivefold higher in the hippocampus compared with the neocortex, our data strongly suggest that estrus cycle-dependent synaptogenesis in the female hippocampus results from cyclic release of GnRH.


Subject(s)
Estradiol/biosynthesis , Gonadotropin-Releasing Hormone/metabolism , Hippocampus/metabolism , Animals , Estrus , Female , Gonadotropin-Releasing Hormone/genetics , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger , Rats , Rats, Wistar
19.
J Biol Chem ; 282(43): 31755-65, 2007 Oct 26.
Article in English | MEDLINE | ID: mdl-17716969

ABSTRACT

Neurofibrillary lesions are characteristic for a group of human diseases, named tauopathies, which are characterized by prominent intracellular accumulations of abnormal filaments formed by the microtubule-associated protein Tau. The tauopathies are accompanied by abnormal changes in Tau protein, including pathological conformation, somatodendritic mislocalization, hyperphosphorylation, and aggregation, whose interdependence is not well understood. To address these issues we have created transgenic mouse lines in which different variants of full-length Tau are expressed in a regulatable fashion, allowing one to switch the expression on and off at defined time points. The Tau variants differ by small mutations in the hexapeptide motifs that control the ability of Tau to adopt a beta-structure conformation and hence to aggregate. The "pro-aggregation" mutant DeltaK280, derived from one of the mutations observed in frontotemporal dementias, aggregates avidly in vitro, whereas the "anti-aggregation" mutant DeltaK280/PP cannot aggregate because of two beta-breaking prolines. In the transgenic mice, the pro-aggregation Tau induces a pathological conformation and pre-tangle aggregation, even at low expression levels, the anti-aggregation mutant does not. This illustrates that abnormal aggregation is primarily controlled by the molecular structure of Tau in vitro and in the organism. Both variants of Tau become mislocalized and hyperphosphorylated independently of aggregation, suggesting that localization and phosphorylation are mainly a consequence of increased concentration. These pathological changes are reversible when the expression of Tau is switched off. The pro-aggregation Tau causes a strong reduction in spine synapses.


Subject(s)
Disease Models, Animal , Synapses/pathology , Tauopathies/pathology , tau Proteins/chemistry , tau Proteins/metabolism , Amino Acid Motifs , Animals , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Mutation , Phosphorylation , Protein Conformation , Protein Isoforms , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/genetics
20.
Exp Neurol ; 203(1): 72-81, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17005180

ABSTRACT

Estrogen has been suggested to be pro-epileptic by reducing GABA synthesis, resulting in increased spine density and a decreased threshold for seizures in the hippocampus, which, once they occur, are characterized by a dramatic spine loss in the affected brain areas. As considerable amounts of estradiol are synthesized in the hippocampus, in this study we focused on aromatase, the rate-limiting enzyme in estrogen synthesis in order to examine the role of locally synthesized estrogens in epilepsy. To this end, we first examined the effects of letrozole, a potent aromatase inhibitor, on GABA metabolism in single interneurons of hippocampal dispersion cultures. Letrozole downregulated estradiol release into the medium, as well as glutamate decarboxylase (GAD) expression and GABA synthesis, and decreased the number of GAD positive cells in the cultures. Next, we counted spine synapses and measured estradiol release of hippocampal slice cultures, in which GABA(A) receptors had been blocked by bicuculline, in order to mimic epileptic activity. Treatment of slice cultures with bicuculline resulted in a dramatic decrease in the number of spine synapses and in a significant suppression of estrogen synthesis. The decrease in synapse number in response to bicuculline was restored by combined application of estradiol and bicuculline. Surprisingly, estradiol alone had no effect on either spine synapse number or on GAD expression and GABA synthesis. "Rescue" of synapse number in "epileptic slices" by estradiol and maintenance of GABA metabolism by hippocampus-derived estradiol points to a neuroprotective role of aromatase in epilepsy. Re-filling of estradiol stores after their depletion due to overexcitation may therefore add to therapeutical strategies in epilepsy.


Subject(s)
Aromatase/metabolism , Dendritic Spines/metabolism , Estradiol/deficiency , Hippocampus/metabolism , Neuroprotective Agents/metabolism , Receptors, GABA-A/metabolism , Animals , Aromatase/drug effects , Cells, Cultured , Dendritic Spines/drug effects , Dendritic Spines/pathology , Down-Regulation/drug effects , Down-Regulation/physiology , Enzyme Inhibitors/pharmacology , Epilepsy/metabolism , Epilepsy/pathology , Epilepsy/physiopathology , Estradiol/biosynthesis , Estradiol/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Glutamate Decarboxylase/drug effects , Glutamate Decarboxylase/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Letrozole , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neuroprotective Agents/pharmacology , Nitriles/pharmacology , Organ Culture Techniques , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Triazoles/pharmacology , gamma-Aminobutyric Acid/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...