Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Biology (Basel) ; 12(4)2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37106832

ABSTRACT

Estrogens via estrogen receptor alpha (ERα) genomic and nongenomic signaling can influence plasticity processes in numerous brain regions. Using mice that express nuclear only ERα (NOER) or membrane only ERα (MOER), this study examined the effect of receptor compartmentalization on the paraventricular nucleus of the hypothalamus (PVN) and the hippocampus. The absence of nuclear and membrane ERα expression impacted females but not males in these two brain areas. In the PVN, quantitative immunohistochemistry showed that the absence of nuclear ERα increased nuclear ERß. Moreover, in the hippocampus CA1, immuno-electron microscopy revealed that the absence of either nuclear or membrane ERα decreased extranuclear ERα and pTrkB in synapses. In contrast, in the dentate gyrus, the absence of nuclear ERα increased pTrkB in synapses, whereas the absence of membrane ERα decreased pTrkB in axons. However, the absence of membrane only ERα decreased the sprouting of mossy fibers in CA3 as reflected by changes in zinc transporter immunolabeling. Altogether these findings support the idea that both membrane and nuclear ERα contribute overlapping and unique actions of estrogen that are tissue- and cellular-specific.

2.
J Neurosci ; 41(24): 5190-5205, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33941651

ABSTRACT

Hypertension susceptibility in women increases at the transition to menopause, termed perimenopause, a state characterized by erratic estrogen fluctuation and extended hormone cycles. Elucidating the role of estrogen signaling in the emergence of hypertension during perimenopause has been hindered by animal models that are confounded by abrupt estrogen cessation or effects of aging. In the present study, accelerated ovarian failure (AOF) in estrogen receptor ß (ERß) reporter mice was induced by 4-vinylcyclohexene diepoxide in young mice to model early-stage ovarian failure (peri-AOF) characteristic of peri-menopause. It was found that administering ERß agonists suppressed elevated blood pressure in a model of neurogenic hypertension induced by angiotensin II (AngII) in peri-AOF, but not in age-matched male mice. It was also found that ERß agonist administration in peri-AOF females, but not males, suppressed the heightened NMDAR signaling and reactive oxygen production in ERß neurons in the hypothalamic paraventricular nucleus (PVN), a critical neural regulator of blood pressure. It was further shown that deleting ERß in the PVN of gonadally intact females produced a phenotype marked by a sensitivity to AngII hypertension. These results suggest that ERß signaling in the PVN plays an important role in blood pressure regulation in female mice and contributes to hypertension susceptibility in females at an early stage of ovarian failure comparable to human perimenopause.


Subject(s)
Estrogen Receptor beta/metabolism , Hypertension/metabolism , Neuronal Plasticity/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Perimenopause/metabolism , Animals , Disease Models, Animal , Female , Hypertension/etiology , Mice , Mice, Inbred C57BL
3.
J Comp Neurol ; 529(9): 2283-2310, 2021 06.
Article in English | MEDLINE | ID: mdl-33341960

ABSTRACT

Within the hypothalamic paraventricular nucleus (PVN), estrogen receptor (ER) ß and other gonadal hormone receptors play a role in central cardiovascular processes. However, the influence of sex and age on the cellular and subcellular relationships of ERß with ERα, G-protein ER (GPER1), as well as progestin and androgen receptors (PR and AR) in the PVN is uncertain. In young (2- to 3-month-old) females and males, ERß-enhanced green fluorescent protein (EGFP) containing neurons were approximately four times greater than ERα-labeled and PR-labeled nuclei in the PVN. In subdivisions of the PVN, young females, compared to males, had: (1) more ERß-EGFP neurons in neuroendocrine rostral regions; (2) fewer ERα-labeled nuclei in neuroendocrine and autonomic projecting medial subregions; and (3) more ERα-labeled nuclei in an autonomic projecting caudal region. In contrast, young males, compared to females, had approximately 20 times more AR-labeled nuclei, which often colocalized with ERß-EGFP in neuroendocrine (approximately 70%) and autonomic (approximately 50%) projecting subregions. Ultrastructurally, in soma and dendrites, PVN ERß-EGFP colocalized primarily with extranuclear AR (approximately 85% soma) and GPER1 (approximately 70% soma). Aged (12- to 24-month-old) males had more ERß-EGFP neurons in a rostral neuroendocrine subregion compared to aged females and females with accelerated ovarian failure (AOF) and in a caudal autonomic subregion compared to post-AOF females. Late-aged (18- to 24-month-old) females compared to early-aged (12- to 14-month-old) females and AOF females had fewer AR-labeled nuclei in neuroendrocrine and autonomic projecting subregions. These findings indicate that gonadal steroids may directly and indirectly influence PVN neurons via nuclear and extranuclear gonadal hormone receptors in a sex-specific manner.


Subject(s)
Estrogen Receptor beta/biosynthesis , Gonadal Steroid Hormones/biosynthesis , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Sex Characteristics , Age Factors , Animals , Estrogen Receptor beta/analysis , Estrogen Receptor beta/ultrastructure , Female , Gonadal Steroid Hormones/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/ultrastructure , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/ultrastructure , Receptors, Androgen/analysis , Receptors, Androgen/biosynthesis , Receptors, Androgen/ultrastructure , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/ultrastructure
4.
Neurobiol Stress ; 13: 100236, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33344692

ABSTRACT

Following oxycodone (Oxy) conditioned place preference (CPP), delta opioid receptors (DORs) differentially redistribute in hippocampal CA3 pyramidal cells in female and male rats in a manner that would promote plasticity and opioid-associative learning processes. However, following chronic immobilization stress (CIS), males do not acquire Oxy-CPP and the trafficking of DORs in CA3 pyramidal neurons is attenuated. Here, we examined the subcellular distribution of DORs in CA1 pyramidal cells using electron microscopy in these same cohorts. CPP: Saline (Sal)-females compared to Sal-males have more cytoplasmic and total DORs in dendrites and more DOR-labeled spines. Following Oxy-CPP, DORs redistribute from near-plasmalemma pools in dendrites to spines in males. CIS: Control females compared to control males have more near-plasmalemmal dendritic DORs. Following CIS, dendritic DORs are elevated in the cytoplasm in females and near-plasmalemma in males. CIS PLUS CPP: CIS Sal-females compared to CIS Sal-males have more DORs on the plasmalemma of dendrites and in spines. After Oxy, the distribution of DORs does not change in either females or males. CONCLUSION: Following Oxy-CPP, DORs within CA1 pyramidal cells remain positioned in naïve female rats to enhance sensitivity to DOR agonists and traffic to dendritic spines in naïve males where they can promote plasticity processes. Following CIS plus behavioral enrichment, DORs are redistributed within CA1 pyramidal cells in females in a manner that could enhance sensitivity to DOR agonists. Conversely, CIS plus behavioral enrichment does not alter DORs in CA1 pyramidal cells in males, which may contribute to their diminished capacity to acquire Oxy-CPP.

5.
Neuroscience ; 423: 192-205, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31682817

ABSTRACT

Sex and ovarian function contribute to hypertension susceptibility, however, the mechanisms are not well understood. Prior studies show that estrogens and neurogenic factors, including hypothalamic glutamatergic NMDA receptor plasticity, play significant roles in rodent hypertension. Here, we investigated the role of sex and ovarian failure on AMPA receptor plasticity in estrogen-sensitive paraventricular nucleus (PVN) neurons in naïve and angiotensin II (AngII) infused male and female mice and female mice at early and late stages of accelerated ovarian failure (AOF). High-resolution electron microscopy was used to assess the subcellular distribution of AMPA GluA1 in age-matched male and female estrogen receptor beta (ERß) enhanced green fluorescent protein (EGFP) reporter mice as well as female ERß-EGFP mice treated with 4-vinylcyclohexene diepoxide. In the absence of AngII, female mice at a late stage of AOF displayed higher levels of GluA1 on the plasma membrane, indicative of functional protein, in ERß-expressing PVN dendrites when compared to male, naïve female and early stage AOF mice. Following slow-pressor AngII infusion, males, as well as early and late stage AOF females had elevated blood pressure. Significantly, only late stage-AOF female mice infused with AngII had an increase in GluA1 near the plasma membrane in dendrites of ERß-expressing PVN neurons. In contrast, prior studies reported that plasmalemmal NMDA GluN1 increased in ERß-expressing PVN dendrites in males and early, but not late stage AOF females. Together, these findings reveal that early and late stage AOF female mice display unique molecular signatures of long-lasting synaptic strength prior to, and following hypertension.


Subject(s)
Cell Membrane/metabolism , Estrogen Receptor beta/metabolism , Hypertension/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Postmenopause/metabolism , Receptors, AMPA/metabolism , Angiotensin II/adverse effects , Animals , Cyclohexenes/adverse effects , Female , Hypertension/chemically induced , Male , Mice , Neurons/metabolism , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/metabolism , Vinyl Compounds/adverse effects
6.
Neurobiol Aging ; 73: 200-210, 2019 01.
Article in English | MEDLINE | ID: mdl-30384123

ABSTRACT

Age and estrogens may impact the mobility of N-methyl-D-aspartate receptors (NMDARs) in hippocampal synapses. Here, we used serial section immunogold electron microscopy to examine whether phosphorylated tyrosine 1472 NR2B (pY1472), which is involved in the surface expression of NMDARs, is altered in the dorsal hippocampus of young (3-4 months old) and aged (∼24 months old) ovariectomized rats treated with 17ß-estradiol or vehicle for 2 days. The number of gold particles labeling pY1472 was higher in presynaptic and postsynaptic compartments of aged rats with low estradiol (vehicle-treated) compared to other groups. In terminals, pY1472 levels were elevated in aged rats but reduced by estradiol treatment to levels seen in young rats. Conversely, the mitochondria number was lower in aged females but was restored to young levels by estradiol. In the postsynaptic density and dendritic spines, estradiol reduced pY1472 in young and aged rats. As phosphorylation at Y1472 blocks NR2B endocytosis, reduction of pY1472 by estradiol suggests another mechanism through which estrogen enhances synaptic plasticity by altering localization of NMDAR subunits within synapses.


Subject(s)
Aging/genetics , Aging/pathology , CA1 Region, Hippocampal/metabolism , Estradiol/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synapses/pathology , Tyrosine/metabolism , Aging/metabolism , Animals , Dendritic Spines , Female , Neuronal Plasticity/drug effects , Ovariectomy , Phosphorylation , Rats, Sprague-Dawley
7.
J Comp Neurol ; 526(14): 2285-2300, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30069875

ABSTRACT

The development of medial temporal lobe circuits is critical for subsequent learning and memory functions later in life. The present study reports the expression of progesterone receptor (PR), a powerful transcription factor of the nuclear steroid receptor superfamily, in Cajal-Retzius cells of the molecular layer of the dentate gyrus of rats. PR was transiently expressed from the day of birth through postnatal day 21, but was absent thereafter. Although PR immunoreactive (PR-ir) cells did not clearly express typical markers of mature neurons, they possessed an ultrastructural morphology consistent with neurons. PRir cells did not express markers for GABAergic neurons, neuronal precursor cells, nor radial glia. However, virtually all PR cells co-expressed the calcium binding protein, calretinin, and the glycoprotein, reelin, both reliable markers for Cajal-Retzius neurons, a transient population of developmentally critical pioneer neurons that guide synaptogenesis of perforant path afferents and histogenesis of the dentate gyrus. Indeed, inhibition of PR activity during the first two weeks of life impaired adult performance on both the novel object recognition and object placement memory tasks, two behavioral tasks hypothesized to describe facets of episodic-like memory in rodents. These findings suggest that PR plays an unexplored and important role in the development of hippocampal circuitry and adult memory function.


Subject(s)
Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Hippocampus/growth & development , Hippocampus/physiology , Memory/physiology , Neurons/metabolism , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/genetics , Animals , Behavior, Animal , Female , Interneurons/metabolism , Memory Disorders/chemically induced , Memory Disorders/genetics , Mifepristone/pharmacology , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/ultrastructure , Pregnancy , Psychomotor Performance/physiology , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/antagonists & inhibitors , Reelin Protein , gamma-Aminobutyric Acid/physiology
8.
Neuroscience ; 383: 98-113, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29753863

ABSTRACT

Corticotropin-releasing factor receptors (CRFR1) contribute to stress-induced adaptations in hippocampal structure and function that can affect learning and memory processes. Our prior studies showed that female rats with elevated estrogens compared to males have more plasmalemmal CRFR1 in CA1 pyramidal cells, suggesting a greater sensitivity to stress. Here, we examined the distribution of hippocampal CRFR1 following chronic immobilization stress (CIS) in female and male rats using immuno-electron microscopy. Without stress, total CRFR1 dendritic levels were higher in females in CA1 and in males in the hilus; moreover, plasmalemmal CRFR1 was elevated in pyramidal cell dendrites in CA1 in females and in CA3 in males. Following CIS, near-plasmalemmal CRFR1 increased in CA1 pyramidal cell dendrites in males but not to levels of control or CIS females. In CA3 and the hilus, CIS decreased cytoplasmic and total CRFR1 in dendrites in males only. These results suggest that in naive rats, CRF could induce a greater activation of CA1 pyramidal cells in females than males. Moreover, after CIS, which leads to even greater sex differences in CRFR1 by trafficking it to different subcellular compartments, CRF could enhance activation of CA1 pyramidal cells in males but to a lesser extent than either unstressed or CIS females. Additionally, CA3 pyramidal cells and inhibitory interneurons in males have heightened sensitivity to CRF, regardless of stress state. These sex differences in CRFR1 distribution and trafficking in the hippocampus may contribute to reported sex differences in hippocampus-dependent learning processes in baseline conditions and following chronic stress.


Subject(s)
Hippocampus/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Sex Characteristics , Stress, Psychological/metabolism , Animals , Female , Male , Rats , Rats, Sprague-Dawley , Restraint, Physical
9.
Nat Commun ; 8(1): 808, 2017 10 09.
Article in English | MEDLINE | ID: mdl-28993643

ABSTRACT

Males and females use distinct brain circuits to cope with similar challenges. Using RNA sequencing of ribosome-bound mRNA from hippocampal CA3 neurons, we found remarkable sex differences and discovered that female mice displayed greater gene expression activation after acute stress than males. Stress-sensitive BDNF Val66Met mice of both sexes show a pre-stressed translational phenotype in which the same genes that are activated without applied stress are also induced in wild-type mice by an acute stressor. Behaviourally, only heterozygous BDNF Val66Met females exhibit spatial memory impairment, regardless of acute stress. Interestingly, this effect is not observed in ovariectomized heterozygous BDNF Val66Met females, suggesting that circulating ovarian hormones induce cognitive impairment in Met carriers. Cognitive deficits are not observed in males of either genotype. Thus, in a brain region not normally associated with sex differences, this work sheds light on ways that genes, environment and sex interact to affect the transcriptome's response to a stressor.Animals' response to acute stress is known to be influenced by sex and genetics. Here the authors performed RNA-seq on actively translated mRNAs in hippocampal CA3 neurons in mice, and document the effects of sex and genotype (i.e., BDNF Val66Met) on acute stress-induced gene expression.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Protein Biosynthesis , Pyramidal Cells/physiology , Stress, Physiological/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Female , Gene Expression Regulation , Glutamic Acid/genetics , Glutamic Acid/metabolism , Male , Mice, Transgenic , Ovariectomy , RNA, Messenger , Sequence Analysis, RNA , Sex Factors , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
10.
Neuroendocrinology ; 104(3): 239-256, 2017.
Article in English | MEDLINE | ID: mdl-27078860

ABSTRACT

Hypertension in male and aging female rodents is associated with glutamate-dependent plasticity in the hypothalamus, but existing models have failed to capture distinct transitional menopausal phases that could have a significant impact on the synaptic plasticity and emergent hypertension. In rodents, accelerated ovarian failure (AOF) induced by systemic injection of 4-vinylcyclohexane diepoxide mimics the estrogen fluctuations seen in human menopause including the perimenopause transition (peri-AOF) and postmenopause (post-AOF). Thus, we used the mouse AOF model to determine the impact of slow-pressor angiotensin II (AngII) administration on blood pressure and on the subcellular distribution of obligatory N-methyl-D-aspartate (NMDA) receptor GluN1 subunits in the paraventricular hypothalamic nucleus (PVN), a key estrogen-responsive cardiovascular regulatory area. Estrogen-sensitive neuronal profiles were identified in mice expressing enhanced green fluorescent protein under the promoter for estrogen receptor (ER) ß, a major ER in the PVN. Slow-pressor AngII increased arterial blood pressure in mice at peri- and post-AOF time points. In control oil-injected (nonhypertensive) mice, AngII decreased the total number of GluN1 in ERß-containing PVN dendrites. In contrast, AngII resulted in a reapportionment of GluN1 from the cytoplasm to the plasma membrane of ERß-containing PVN dendrites in peri-AOF mice. Moreover, in post-AOF mice, AngII increased total GluN1, dendritic size and radical production in ERß-containing neurons. These results indicate that unique patterns of hypothalamic glutamate receptor plasticity and dendritic structure accompany the elevated blood pressure in peri- and post-AOF time points. Our findings suggest the possibility that distinct neurobiological processes are associated with the increased blood pressure during perimenopausal and postmenopausal periods.


Subject(s)
Hypertension , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Ovarian Diseases/etiology , Paraventricular Hypothalamic Nucleus/pathology , Receptors, Estrogen/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Angiotensin II/toxicity , Animals , Blood Pressure/drug effects , Cyclohexenes/toxicity , Disease Models, Animal , Estrous Cycle/drug effects , Estrous Cycle/genetics , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypertension/chemically induced , Hypertension/complications , Hypertension/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Immunoelectron , Neurons/ultrastructure , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/ultrastructure , Reactive Oxygen Species/metabolism , Receptors, Estrogen/genetics , Vinyl Compounds/toxicity
11.
Neurobiol Stress ; 5: 37-53, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27981195

ABSTRACT

Drug addiction requires associative learning processes that critically involve hippocampal circuits, including the opioid system. We recently found that acute and chronic stress, important regulators of addictive processes, affect hippocampal opioid levels and mu opioid receptor trafficking in a sexually dimorphic manner. Here, we examined whether acute and chronic stress similarly alters the levels and trafficking of hippocampal delta opioid receptors (DORs). Immediately after acute immobilization stress (AIS) or one-day after chronic immobilization stress (CIS), the brains of adult female and male rats were perfusion-fixed with aldehydes. The CA3b region and the dentate hilus of the dorsal hippocampus were quantitatively analyzed by light microscopy using DOR immunoperoxidase or dual label electron microscopy for DOR using silver intensified immunogold particles (SIG) and GABA using immunoperoxidase. At baseline, females compared to males had more DORs near the plasmalemma of pyramidal cell dendrites and about 3 times more DOR-labeled CA3 dendritic spines contacted by mossy fibers. In AIS females, near-plasmalemmal DOR-SIGs decreased in GABAergic hilar dendrites. However, in AIS males, near-plasmalemmal DOR-SIGs increased in CA3 pyramidal cell and hilar GABAergic dendrites and the percentage of CA3 dendritic spines contacted by mossy fibers increased to about half that seen in unstressed females. Conversely, after CIS, near-plasmalemmal DOR-SIGs increased in hilar GABA-labeled dendrites of females whereas in males plasmalemmal DOR-SIGs decreased in CA3 pyramidal cell dendrites and near-plasmalemmal DOR-SIGs decreased hilar GABA-labeled dendrites. As CIS in females, but not males, redistributed DOR-SIGs near the plasmalemmal of hilar GABAergic dendrites, a subsequent experiment examined the acute affect of oxycodone on the redistribution of DOR-SIGs in a separate cohort of CIS females. Plasmalemmal DOR-SIGs were significantly elevated on hilar interneuron dendrites one-hour after oxycodone (3 mg/kg, I.P.) administration compared to saline administration in CIS females. These data indicate that DORs redistribute within CA3 pyramidal cells and dentate hilar GABAergic interneurons in a sexually dimorphic manner that would promote activation and drug related learning in males after AIS and in females after CIS.

12.
J Comp Neurol ; 524(11): 2251-65, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26659944

ABSTRACT

At younger ages, women have a lower risk for hypertension than men, but this sexual dimorphism declines with the onset of menopause. These differences are paralleled in rodents following "slow-pressor" angiotensin II (AngII) administration: young male and aged female mice, but not young females, develop hypertension. There is also an established sexual dimorphism both in the cardiovascular response to the neurohypophyseal hormone arginine vasopressin (AVP) and in the expression of oxidative stress. We examined the relationship between AngII-mediated hypertension and the cellular distribution of the superoxide generating NADPH oxidase (NOX) in AVP-expressing hypothalamic paraventricular nucleus (PVN) neurons in "menopausal" female mice. Dual-labeling immunoelectron microscopy was used to determine whether the subcellular distribution of the organizer/adapter NOX p47(phox) subunit is altered in PVN dendrites following AngII administered (14 days) during the "postmenopausal" stage of accelerated ovarian failure (AOF) in young female mice treated with 4-vinylcyclohexene diepoxide. Slow-pressor AngII elevated blood pressure in AOF females and induced a significant increase in near plasmalemmal p47(phox) and a decrease in cytoplasmic p47(phox) in PVN AVP dendrites. These changes are the opposite of those observed in AngII-induced hypertensive male mice (Coleman et al. [2013] J. Neurosci. 33:4308-4316) and may be ascribed in part to baseline differences between young females and males in the near plasmalemmal p47(phox) on AVP dendrites seen in the present study. These findings highlight fundamental differences in the neural substrates of oxidative stress in the PVN associated with AngII hypertension in postmenopausal females compared with males. J. Comp. Neurol. 524:2251-2265, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Hypertension/metabolism , NADPH Oxidases/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Postmenopause/metabolism , Sex Characteristics , Angiotensin II/toxicity , Animals , Disease Models, Animal , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Neurons/enzymology
13.
Physiol Rev ; 95(3): 785-807, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26109339

ABSTRACT

Estrogen facilitates higher cognitive functions by exerting effects on brain regions such as the prefrontal cortex and hippocampus. Estrogen induces spinogenesis and synaptogenesis in these two brain regions and also initiates a complex set of signal transduction pathways via estrogen receptors (ERs). Along with the classical genomic effects mediated by activation of ER α and ER ß, there are membrane-bound ER α, ER ß, and G protein-coupled estrogen receptor 1 (GPER1) that can mediate rapid nongenomic effects. All key ERs present throughout the body are also present in synapses of the hippocampus and prefrontal cortex. This review summarizes estrogen actions in the brain from the standpoint of their effects on synapse structure and function, noting also the synergistic role of progesterone. We first begin with a review of ER subtypes in the brain and how their abundance and distributions are altered with aging and estrogen loss (e.g., ovariectomy or menopause) in the rodent, monkey, and human brain. As there is much evidence that estrogen loss induced by menopause can exacerbate the effects of aging on cognitive functions, we then review the clinical trials of hormone replacement therapies and their effectiveness on cognitive symptoms experienced by women. Finally, we summarize studies carried out in nonhuman primate models of age- and menopause-related cognitive decline that are highly relevant for developing effective interventions for menopausal women. Together, we highlight a new understanding of how estrogen affects higher cognitive functions and synaptic health that go well beyond its effects on reproduction.


Subject(s)
Aging/metabolism , Aging/psychology , Brain/metabolism , Cognition , Estrogens/metabolism , Synapses/metabolism , Synaptic Transmission , Age Factors , Animals , Cognition Disorders/metabolism , Cognition Disorders/psychology , Female , Humans , Male , Menopause/metabolism , Menopause/psychology , Progesterone/metabolism , Receptors, Estrogen/metabolism
14.
J Neurosci ; 35(6): 2384-97, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673833

ABSTRACT

Both estrous cycle and sex affect the numbers and types of neuronal and glial profiles containing the classical estrogen receptors α and ß, and synaptic levels in the rodent dorsal hippocampus. Here, we examined whether the membrane estrogen receptor, G-protein-coupled estrogen receptor 1 (GPER1), is anatomically positioned in the dorsal hippocampus of mice to regulate synaptic plasticity. By light microscopy, GPER1-immunoreactivity (IR) was most noticeable in the pyramidal cell layer and interspersed interneurons, especially those in the hilus of the dentate gyrus. Diffuse GPER1-IR was found in all lamina but was most dense in stratum lucidum of CA3. Ultrastructural analysis revealed discrete extranuclear GPER1-IR affiliated with the plasma membrane and endoplasmic reticulum of neuronal perikarya and dendritic shafts, synaptic specializations in dendritic spines, and clusters of vesicles in axon terminals. Moreover, GPER1-IR was found in unmyelinated axons and glial profiles. Overall, the types and amounts of GPER1-labeled profiles were similar between males and females; however, in females elevated estrogen levels generally increased axonal labeling. Some estradiol-induced changes observed in previous studies were replicated by the GPER agonist G1: G1 increased PSD95-IR in strata oriens, lucidum, and radiatum of CA3 in ovariectomized mice 6 h after administration. In contrast, estradiol but not G1 increased Akt phosphorylation levels. Instead, GPER1 actions in the synapse may be due to interactions with synaptic scaffolding proteins, such as SAP97. These results suggest that although estrogen's actions via GPER1 may converge on the same synaptic elements, different pathways are used to achieve these actions.


Subject(s)
Hippocampus/physiology , Hippocampus/ultrastructure , Neuronal Plasticity/physiology , Receptors, G-Protein-Coupled/metabolism , Synapses/physiology , Synapses/ultrastructure , Animals , Discs Large Homolog 1 Protein , Disks Large Homolog 4 Protein , Estrous Cycle/physiology , Female , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Hippocampus/drug effects , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, Presynaptic/metabolism , Receptors, Presynaptic/ultrastructure , Sex Characteristics , Synapses/drug effects
15.
Synapse ; 69(3): 148-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25559190

ABSTRACT

Renin­angiotensin system overactivity, upregulation of postsynaptic NMDA receptor function, and increased reactive oxygen species (ROS) production in the hypothalamic paraventricular nucleus (PVN) are hallmarks of angiotensin II (AngII)-induced hypertension, which is far more common in young males than in young females. We hypothesize that the sex differences in hypertension are related to differential AngII-induced changes in postsynaptic trafficking of the essential NMDA receptor GluN1 subunit and ROS production in PVN cells expressing angiotensin Type 1a receptor (AT1aR). We tested this hypothesis using slow-pressor (14-day) infusion of AngII (600 ng/kg/min) in mice, which elicits hypertension in males but not in young females. Two-month-old male and female transgenic mice expressing enhanced green fluorescent protein (EGFP) in AT1aR-containing cells were used. In males, but not in females, AngII increased blood pressure and ROS production in AT1aR­EGFP PVN cells at baseline and following NMDA treatment. Electron microscopy showed that AngII increased cytoplasmic and total GluN1­silver-intensified immunogold (SIG) densities and induced a trend toward an increase in near plasmalemmal GluN1­SIG density in AT1aR­EGFP dendrites of males and females. Moreover, AngII decreased dendritic area and diameter in males, but increased dendritic area of small (<1 µm) dendrites and decreased diameter of large (>1 µm) dendrites in females. Fluorescence microscopy revealed that AT1aR and estrogen receptor ß do not colocalize, suggesting that if estrogen is involved, its effect is indirect. These data suggest that the sexual dimorphism in AngII-induced hypertension is associated with sex differences in ROS production in AT1aR-containing PVN cells but not with postsynaptic NMDA receptor trafficking.


Subject(s)
Angiotensin II/pharmacology , Dendrites/metabolism , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Blood Pressure , Dendrites/ultrastructure , Estrogen Receptor beta/metabolism , Female , Hypothalamus/cytology , Hypothalamus/drug effects , Male , Mice , Protein Transport , Receptor, Angiotensin, Type 1/genetics , Renin-Angiotensin System , Sex Factors
16.
Endocrinology ; 155(9): 3610-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24926825

ABSTRACT

Accelerated ovarian failure (AOF) can be induced in young mice with low doses of 4-vinylcyclohexene diepoxide (VCD), modeling the hormone changes observed across menopause. We assessed markers of synaptic plasticity in the hippocampus, anxiety-like behavior, and spatial learning longitudinally at 4 time points across the AOF model: premenopause, early perimenopause, late perimenopause, and postmenopause (POST). As others have shown, VCD administration decreased ovarian follicle counts and increased acyclicity as the model progressed to POST but with no impact on organ or body weights. The morphology of Iba1 immunoreactive microglia did not differ between vehicle- and VCD-administered mice. Hippocampal postsynaptic density 95 levels were minimally altered across the AOF model but decreased at POST in CA3b 24 hours after exogenous estradiol benzoate (EB). In contrast, hippocampal phosphorylated AKT levels transiently decreased in premenopause but increased at POST after 24 hours of EB in select subregions. Electron microscopy revealed fewer estrogen receptor α containing dendritic spines and terminals in CA1 stratum radiatum at POST. mRNA levels of most brain-derived neurotrophic factor exons (except V and VI) were lower in POST compared with ovariectomized mice. Exon V was sensitive to 24 hours of EB administration in POST-VCD. Anxiety-like behavior was unaffected at any menopause phase. Spatial learning was unaffected in all groups, but POST-VCD mice performed below chance. Our results suggest that the AOF model is suitable for longitudinal studies of neurobiological changes across the menopause transition in mice. Our findings also point to complex interactions between estrogen receptors and pathways involved in synaptic plasticity.


Subject(s)
Estrogens/metabolism , Menopause/metabolism , Nerve Growth Factors/metabolism , Primary Ovarian Insufficiency/metabolism , Signal Transduction , Animals , Disease Models, Animal , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Hippocampus/metabolism , Humans , Menopause/genetics , Mice , Nerve Growth Factors/genetics , Primary Ovarian Insufficiency/genetics
17.
J Comp Neurol ; 522(13): 3075-90, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24639345

ABSTRACT

The incidence of hypertension increases after menopause. Similar to humans, "slow-pressor" doses of angiotensin II (AngII) increase blood pressure in young males, but not in young female mice. However, AngII increases blood pressure in aged female mice, paralleling reproductive hormonal changes. These changes could influence receptor trafficking in central cardiovascular circuits and contribute to hypertension. Increased postsynaptic N-methyl-D-aspartate (NMDA) receptor activity in the hypothalamic paraventricular nucleus (PVN) is crucial for the sympathoexcitation driving AngII hypertension. Estrogen receptors ß (ERßs) are present in PVN neurons. We tested the hypothesis that changes in ovarian hormones with age promote susceptibility to AngII hypertension, and influence NMDA receptor NR1 subunit trafficking in ERß-containing PVN neurons. Transgenic mice expressing enhanced green fluorescent protein (EGFP) in ERß-containing cells were implanted with osmotic minipumps delivering AngII (600 ng/kg/min) or saline for 2 weeks. AngII increased blood pressure in 2-month-old males and 18-month-old females, but not in 2-month-old females. By electron microscopy, NR1-silver-intensified immunogold (SIG) was mainly in ERß-EGFP dendrites. At baseline, NR1-SIG density was greater in 2-month-old females than in 2-month-old males or 18-month-old females. After AngII infusion, NR1-SIG density was decreased in 2-month-old females, but increased in 2-month-old males and 18-month-old females. These findings suggest that, in young female mice, NR1 density is decreased in ERß-PVN dendrites thus reducing NMDA receptor activity and preventing hypertension. Conversely, in young males and aged females, NR1 density is upregulated in ERß-PVN dendrites and ultimately leads to the neurohumoral dysfunction driving hypertension.


Subject(s)
Aging/physiology , Angiotensin II/administration & dosage , Dendrites/metabolism , Estrogen Receptor beta/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Blood Pressure/drug effects , Estrogen Receptor beta/genetics , Estrous Cycle/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Neurons/ultrastructure , Sex Factors , Stilbamidines/metabolism , Time Factors
18.
J Chem Neuroanat ; 55: 9-17, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24275289

ABSTRACT

Research indicates that responses to stress are sexually dimorphic, particularly in regard to learning and memory processes: while males display impaired cognitive performance and hippocampal CA3 pyramidal cell dendritic remodeling following chronic stress, females exhibit enhanced performance and no remodeling. Leu-enkephalin, an endogenous opioid peptide found in the hippocampal mossy fiber pathway, plays a critical role in mediating synaptic plasticity at the mossy fiber-CA3 pyramidal cell synapse. Estrogen is known to influence the expression of leu-enkephalin in the mossy fibers of females, with leu-enkephalin levels being highest at proestrus and estrus, when estrogen levels are elevated. Since stress is also known to alter the expression of leu-enkephalin in various brain regions, this study was designed to determine whether acute or chronic stress had an effect on mossy fiber leu-enkephalin levels in females or males, through the application of correlated quantitative light and electron microscopic immunocytochemistry. Both acute and chronic stress eliminated the estrogen-dependence of leu-enkephalin levels across the estrous cycle in females, but had no effect on male levels. However, following acute stress leu-enkephalin levels in females were consistently lowered to values comparable to the lowest control values, while following chronic stress they were consistently elevated to values comparable to the highest control values. Ultrastructural changes in leu-enkephalin labeled dense core vesicles paralleled light microscopic observations, with acute stress inducing a decrease in leu-enkephalin labeled dense core vesicles, and chronic stress inducing an increase in leu-enkephalin labeled dense-core vesicles in females. These findings suggest that alterations in leu-enkephalin levels following stress could play an important role in the sex-specific responses that females display in learning processes, including those important in addiction.


Subject(s)
Enkephalin, Leucine/metabolism , Mossy Fibers, Hippocampal/metabolism , Stress, Physiological/physiology , Stress, Psychological/metabolism , Animals , Dendrites/metabolism , Estrous Cycle/metabolism , Female , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Sex Factors
19.
Neurosci Lett ; 552: 66-70, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23933204

ABSTRACT

The opioid peptides, dynorphin (DYN) and enkephalin (L-ENK) are contained in the hippocampal mossy fiber pathway where they modulate synaptic plasticity. In rats, the levels of DYN and L-ENK immunoreactivity (-ir) are increased when estrogen levels are elevated (Torres-Reveron et al., 2008, 2009). Here, we used quantitative immunocytochemistry to examine whether opioid levels are similarly regulated in wildtype (WT) mice over the estrous cycle, and how these compared to males. Moreover, using estrogen receptor (ER) alpha and beta knock-out mice (AERKO and BERKO, respectively), the present study examined the role of ERs in rapid, membrane-initiated (6 h), or slower, nucleus-initiated (48 h) estradiol effects on mossy fiber opioid levels. Unlike rats, the levels of DYN and L-ENK-ir did not change over the estrous cycle. However, compared to males, females had higher levels of DYN-ir in CA3a and L-ENK-ir in CA3b. In WT and BERKO ovariectomized (OVX) mice, neither DYN- nor L-ENK-ir changed following 6 or 48 h estradiol benzoate (EB) administration. However, DYN-ir significantly increased 48 h after EB in the dentate gyrus (DG) and CA3b of AERKO mice only. These findings suggest that cyclic hormone levels regulate neither DYN nor L-ENK levels in the mouse mossy fiber pathway as they do in the rat. This may be due to species-specific differences in the mossy fiber pathway. However, in the mouse, DYN levels are regulated by exogenous EB in the absence of ERα possibly via an ERß-mediated pathway requiring new gene transcription.


Subject(s)
CA3 Region, Hippocampal/metabolism , Dynorphins/metabolism , Enkephalins/metabolism , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Mossy Fibers, Hippocampal/metabolism , Animals , CA3 Region, Hippocampal/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrous Cycle/metabolism , Female , Male , Mice , Mice, Knockout , Mossy Fibers, Hippocampal/drug effects , Sex Characteristics
20.
Synapse ; 67(11): 757-72, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23720407

ABSTRACT

Stress differentially affects hippocampal-dependent learning relevant to addiction and morphology in male and female rats. Mu opioid receptors (MORs), which are located in parvalbumin (PARV)-containing GABAergic interneurons and are trafficked in response to changes in the hormonal environment, play a critical role in promoting principal cell excitability and long-term potentiation. Here, we compared the effects of acute and chronic immobilization stress (AIS and CIS) on MOR trafficking in PARV-containing neurons in the hilus of the dentate gyrus in female and male rats using dual label immunoelectron microscopy. Following AIS, the density of MOR silver-intensified gold particles (SIGs) in the cytoplasm of PARV-labeled dendrites was significantly reduced in females (estrus stage). Conversely, AIS significantly increased the proportion of cytoplasmic MOR SIGs in PARV-labeled dendrites in male rats. CIS significantly reduced the number of PARV-labeled neurons in the dentate hilus of males but not females. However, MOR/PARV-labeled dendrites and terminals were significantly smaller in CIS females, but not males, compared with controls. Following CIS, the density of cytoplasmic MOR SIGs increased in PARV-labeled dendrites and terminals in females. Moreover, the proportion of near-plasmalemmal MOR SIGs relative to total decreased in large PARV-labeled dendrites in females. After CIS, no changes in the density or trafficking of MOR SIGs were seen in PARV-labeled dendrites or terminals in males. These data show that AIS and CIS differentially affect available MOR pools in PARV-containing interneurons in female and male rats. Furthermore, they suggest that CIS could affect principal cell excitability in a manner that maintains learning processes in females but not males.


Subject(s)
Dentate Gyrus/metabolism , Interneurons/metabolism , Parvalbumins/analysis , Receptors, Opioid, mu/metabolism , Stress, Psychological/metabolism , Animals , Cell Membrane/metabolism , Cytoplasm/metabolism , Dendrites/metabolism , Dentate Gyrus/cytology , Female , Interneurons/chemistry , Male , Protein Transport , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/genetics , Sex Characteristics
SELECTION OF CITATIONS
SEARCH DETAIL
...