Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Open Biol ; 13(11): 230217, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37989220

ABSTRACT

Pregnancy is marked by robust changes, including brain changes to volume, structure, connectivity and neuroplasticity. Although some brain changes are restricted to pregnancy and the postpartum, others are long-lasting. Few studies have examined possible mechanisms of these changes or the effects of multiple pregnancies. We characterized various cellular and molecular signatures of parity (nulliparous, primiparous, biparous) in the rat hippocampus. We investigated density of neural stems cells (Sox2), microglia (Iba-1) and levels of a synaptic protein (PSD-95), cell signalling pathways, neuroinflammation, and the tryptophan-kynurenine (TRP-KYN) pathway, one week after weaning their pups from the last pregnancy (age of dam: seven months) and in middle-age (age of dam: 13 months). Parity increased PSD-95 levels in both age groups and prevented the age-related decrease in neural stem cell density observed in nulliparous rats. Biparity increased cell signalling phosphoproteins (pp70S6K, S6RP) and number of microglia in the dentate gyrus, regardless of age. Parity resulted in transient changes to the TRP-KYN system. Thus, previous parity has lasting effects on synaptic plasticity with fewer lasting effects on inflammation and cell signalling phosphoproteins in the whole hippocampus.


Subject(s)
Brain , Tryptophan , Pregnancy , Humans , Female , Rats , Animals , Tryptophan/metabolism , Brain/metabolism , Kynurenine/metabolism , Postpartum Period , Phosphoproteins/metabolism
2.
J Neuroendocrinol ; 30(4): e12578, 2018 04.
Article in English | MEDLINE | ID: mdl-29411916

ABSTRACT

Adult hippocampal neurogenesis occurs in many mammalian species. In rats, the survival of new neurones within the hippocampus is modulated by the action of androgen via the androgen receptor (AR); however, it is not known whether this holds true in mice. Furthermore, the evidence is mixed regarding whether androgens act in neural tissue or via peripheral non-neural targets to promote new neurone survival in the hippocampus. We evaluated whether the action of androgen via AR underlies the survival of new neurones in mice, and investigated whether increasing AR selectively in neural tissue would increase new neurone survival in the hippocampus. We used the cre-loxP system to overexpress AR only in neural tissues (Nestin-AR). These males were compared with wild-type males, as well as control males with 1 of the 2 mutations required for overexpression. Mice were gonadectomised and injected with the DNA synthesis marker, bromodeoxyuridine (BrdU) and for 37 days (following BrdU injection), mice were treated with oil or dihydrotestosterone (DHT). Using immunohistochemistry, proliferation (Ki67) and survival (BrdU) of new neurones were both evaluated in the dorsal and ventral dentate gyrus. Dihydrotestosterone treatment increased the survival of new neurones in the entire hippocampus in wild-type mice and control mice that only have 1 of 2 necessary mutations for transgenic expression. However, DHT treatment did not increase the survival of new neurones in mice that overexpressed AR in neural tissue. Cell proliferation (Ki67) and cell death (pyknotic cells) were not affected by DHT treatment in wild-type or transgenic males. These results suggest that androgens act via neural AR to affect hippocampal neurogenesis by promoting cell survival; however, the relationship between androgen dose and new neurone survival is nonlinear.


Subject(s)
Cell Survival/physiology , Dentate Gyrus/metabolism , Neurons/metabolism , Receptors, Androgen/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dihydrotestosterone/pharmacology , Male , Mice , Mice, Transgenic , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/cytology , Neurons/drug effects , Receptors, Androgen/genetics
3.
J Neuroendocrinol ; 27(10): 772-86, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26258544

ABSTRACT

Gonadotrophin-releasing hormone (GnRH) and gonadotrophin inhibitory hormone (GnIH) are neuropeptides secreted by the hypothalamus that regulate reproduction. GnRH receptors are not only present in the anterior pituitary, but also are abundantly expressed in the hippocampus of rats, suggesting that GnRH regulates hippocampal function. GnIH inhibits pituitary gonadotrophin secretion and is also expressed in the hippocampus of a songbird; its role outside of the reproductive axis is not well established. In the present study, we employed immunohistochemistry to examine three forms of GnRH [mammalian GnRH-I (mGnRH-I), chicken GnRH-II (cGnRH-II) and lamprey GnRH-III (lGnRH-III)] and GnIH in the adult rat hippocampus. No mGnRH-I and cGnRH-II+ cell bodies were present in the hippocampus. Sparse mGnRH-I and cGnRH-II+ fibres were present within the CA1 and CA3 fields of the hippocampus, along the hippocampal fissure, and within the hilus of the dentate gyrus. No lGnRH-III was present in the rodent hippocampus. GnIH-immunoreactivity was present in the hippocampus in cell bodies that resembled astrocytes. Males had more GnIH+ cells in the hilus of the dentate gyrus than females. To confirm the GnIH+ cell body phenotype, we performed double-label immunofluorescence against GnIH, glial fibrillary acidic protein (GFAP) and NeuN. Immunofluorescence revealed that all GnIH+ cell bodies in the hippocampus also contained GFAP, a marker of astrocytes. Taken together, these data suggest that GnRH does not reach GnRH receptors in the rat hippocampus primarily via synaptic release. By contrast, GnIH might be synthesised locally in the rat hippocampus by astrocytes. These data shed light on the sites of action and possible functions of GnRH and GnIH outside of the hypothalamic-pituitary-gonadal axis.


Subject(s)
Astrocytes/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hippocampus/metabolism , Hypothalamic Hormones/physiology , Neurons/metabolism , Animals , Female , Male , Rats, Long-Evans
4.
J Neuroendocrinol ; 26(10): 641-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25039797

ABSTRACT

The peripartum period is accompanied by dramatic changes in hormones and a host of new behaviours in response to experience with offspring. Both maternal experience and maternal hormones can have a significant impact upon the brain and behaviour. This review outlines recent studies demonstrating modifications in hippocampal plasticity across the peripartum period, as well as the putative hormonal mechanisms underlying these changes and their modulation by stress. In addition, the impact of reproductive experience upon the ageing hippocampus is discussed. Finally, we consider how these changes in hippocampal structure may play a role in postpartum cognitive function and mood disorders, as well as age-related cognitive decline.


Subject(s)
Aging/physiology , Gonadal Steroid Hormones/physiology , Hippocampus/physiology , Neuronal Plasticity , Stress, Physiological , Animals , Cognition , Female , Models, Animal , Neurogenesis , Pregnancy
5.
J Neuroendocrinol ; 26(6): 386-99, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24750490

ABSTRACT

Both natural oestrogens and progesterone influence synaptic plasticity and neurogenesis within the female hippocampus. However, less is known of the impact of synthetic hormones on hippocampal structure and function. There is some evidence that the administration of the synthetic progestin, medroxyprogesterone acetate (MPA) is not as beneficial as natural progesterone and can attenuate oestrogen-induced neuroprotection. Although the effects of oestradiol have been well studied, little is known about the effects of natural and synthetic progestins alone and in combination with oestradiol on adult neurogenesis in females. In the present study, we investigated the effects of chronic oestradiol, progesterone, MPA and the co-administration of each progestin with oestradiol on neurogenesis within the dentate gyrus of adult ovariectomised female rats. Twenty-four hours after a bromodeoxyuridine (BrdU; 200 mg/kg) injection, female rats were repeatedly administered either progesterone (1 or 4 mg), MPA (1 or 4 mg), oestradiol benzoate (EB), progesterone or MPA in combination with EB (10 µg), or vehicle for 21 days. Rats were perfused on day 22 and brain tissue was analysed for the number of BrdU-labelled and Ki67 (an endogenous marker of cell proliferation)-expressing cells. EB alone and MPA + EB significantly decreased neurogenesis and the number of surviving BrdU-labelled cells in the dorsal region of the dentate gyrus, independent of any effects on cell proliferation. Furthermore, MPA (1 and 4 mg) and MPA + EB treated animals had significantly lower adrenal/body mass ratios and reduced serum corticosterone (CORT) levels. By contrast, progesterone + EB treated animals had significantly higher adrenal/body mass ratios and 1 mg of progesterone, progesterone + EB, and EB significantly increased CORT levels. The results of the present study demonstrate that different progestins alone and in combination with oestradiol can differentially affect neurogenesis (via cell survival) and regulation of the hypothalamic-pituitary-adrenal axis. These findings have implications for women using hormone replacement therapies with MPA for both neuroprotection and stress-related disorders.


Subject(s)
Adrenal Glands/growth & development , Contraceptives, Oral, Hormonal/pharmacology , Estradiol/pharmacology , Estrogens/pharmacology , Hippocampus/growth & development , Medroxyprogesterone/pharmacology , Neurogenesis/drug effects , Progesterone/pharmacology , Adrenal Glands/drug effects , Animals , Antigens, Nuclear/biosynthesis , Dentate Gyrus/drug effects , Dentate Gyrus/growth & development , Female , Hippocampus/cytology , Hippocampus/drug effects , Ki-67 Antigen/biosynthesis , Nerve Tissue Proteins/biosynthesis , Rats , Rats, Sprague-Dawley
6.
J Neuroendocrinol ; 25(11): 1039-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23822747

ABSTRACT

The hippocampus is an area of the brain that undergoes dramatic plasticity in response to experience and hormone exposure. The hippocampus retains the ability to produce new neurones in most mammalian species and is a structure that is targeted in a number of neurodegenerative and neuropsychiatric diseases, many of which are influenced by both sex and sex hormone exposure. Intriguingly, gonadal and adrenal hormones affect the structure and function of the hippocampus differently in males and females. Adult neurogenesis in the hippocampus is regulated by both gonadal and adrenal hormones in a sex- and experience-dependent way. Sex differences in the effects of steroid hormones to modulate hippocampal plasticity should not be completely unexpected because the physiology of males and females is different, with the most notable difference being that females gestate and nurse the offspring. Furthermore, reproductive experience (i.e. pregnancy and mothering) results in permanent changes to the maternal brain, including the hippocampus. This review outlines the ability of gonadal and stress hormones to modulate multiple aspects of neurogenesis (cell proliferation and cell survival) in both male and female rodents. The function of adult neurogenesis in the hippocampus is linked to spatial memory and depression, and the present review provides early evidence of the functional links between the hormonal modulation of neurogenesis that may contribute to the regulation of cognition and stress.


Subject(s)
Cognition/physiology , Hippocampus/physiology , Hormones/physiology , Neurogenesis/physiology , Rodentia/physiology , Sex Characteristics , Animals , Female , Hippocampus/metabolism , Hormones/metabolism , Male , Rodentia/metabolism
7.
Endocrinology ; 154(9): 3294-304, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23782943

ABSTRACT

Gonadal steroids are potent regulators of adult neurogenesis. We previously reported that androgens, such as testosterone (T) and dihydrotestosterone (DHT), but not estradiol, increased the survival of new neurons in the dentate gyrus of the male rat. These results suggest androgens regulate hippocampal neurogenesis via the androgen receptor (AR). To test this supposition, we examined the role of ARs in hippocampal neurogenesis using 2 different approaches. In experiment 1, we examined neurogenesis in male rats insensitive to androgens due to a naturally occurring mutation in the gene encoding the AR (termed testicular feminization mutation) compared with wild-type males. In experiment 2, we injected the AR antagonist, flutamide, into castrated male rats and compared neurogenesis levels in the dentate gyrus of DHT and oil-treated controls. In experiment 1, chronic T increased hippocampal neurogenesis in wild-type males but not in androgen-insensitive testicular feminization mutation males. In experiment 2, DHT increased hippocampal neurogenesis via cell survival, an effect that was blocked by concurrent treatment with flutamide. DHT, however, did not affect cell proliferation. Interestingly, cells expressing doublecortin, a marker of immature neurons, did not colabel with ARs in the dentate gyrus, but ARs were robustly expressed in other regions of the hippocampus. Together these studies provide complementary evidence that androgens regulate adult neurogenesis in the hippocampus via the AR but at a site other than the dentate gyrus. Understanding where in the brain androgens act to increase the survival of new neurons in the adult brain may have implications for neurodegenerative disorders.


Subject(s)
Androgens/metabolism , Dentate Gyrus/metabolism , Neurogenesis , Neurons/metabolism , Neuroprotective Agents/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Androgen Antagonists/toxicity , Androgen-Insensitivity Syndrome/chemically induced , Androgen-Insensitivity Syndrome/drug therapy , Androgen-Insensitivity Syndrome/metabolism , Androgens/chemistry , Androgens/pharmacology , Androgens/therapeutic use , Animals , Biomarkers/metabolism , Castration/adverse effects , Cell Survival/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Doublecortin Domain Proteins , Doublecortin Protein , Drug Resistance , Hormone Replacement Therapy , Male , Microtubule-Associated Proteins/metabolism , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Neuropeptides/metabolism , Neuroprotective Agents/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Signal Transduction/drug effects , Testosterone Propionate/antagonists & inhibitors , Testosterone Propionate/pharmacology , Testosterone Propionate/therapeutic use
8.
J Neuroendocrinol ; 25(2): 119-30, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22935038

ABSTRACT

Postpartum depression (PPD) affects approximately 15% of mothers after giving birth. A complete understanding of depression during the postpartum period has yet to be established, although disruptions in the hypothalamic-pituitary-adrenal axis and stress during the postpartum may be involved. To model these components in rats, we administered high corticosterone (CORT) postpartum, which increases immobility in the forced swim test (FST), and reduces maternal care, body weight and hippocampal cell proliferation in dams. The hippocampus is altered in response to chronic stress, exposure to high glucocorticoids and in major depression in humans. In the present study, we examined whether high CORT reduced dendritic complexity and spines in the CA3 region of the hippocampus. Additionally, housing complexity was manipulated so that dams and litters were housed either with tubes (complex) or without tubes (impoverished) to investigate the consequences of new animal care regulations. Dams received 40 mg/kg/day of CORT or oil starting on day 2 postpartum for 23 days. Maternal behaviours were assessed on postpartum days 2-8 and dams were tested using the FST on days 21 and 22. Dams were killed on day 24 and brains were processed for Golgi impregnation. Pyramidal cells in the CA3 subfield were traced using a camera lucida and analysed for branch points and dendritic complexity, as well as spine density and type on both basal and apical arbours. As previously established, high CORT postpartum reduced maternal care and increased immobility in the FST, which is a measure of depressive-like behaviour. High CORT postpartum reduced the complexity of basal arbours and increased mushroom spines on both apical and basal dendrites. Housing complexity had no effect on spines of CA3 pyramidal cells but modest effects on cell morphology. These data show that chronic high CORT in postpartum females alters hippocampal morphology and may provide insight regarding the neurobiological consequences of high stress or CORT during the postpartum period, as well as be relevant for postpartum stress or depression.


Subject(s)
Corticosterone/metabolism , Dendritic Spines/pathology , Depression, Postpartum/metabolism , Hippocampus/pathology , Postpartum Period/metabolism , Animals , Cell Count , Cell Proliferation/drug effects , Corticosterone/administration & dosage , Dendrites/drug effects , Dendrites/pathology , Dendritic Spines/drug effects , Depression, Postpartum/pathology , Down-Regulation/drug effects , Female , Hippocampus/drug effects , Male , Rats , Rats, Sprague-Dawley , Time Factors
9.
Dev Psychobiol ; 55(5): 518-29, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22692766

ABSTRACT

This study was undertaken to determine whether altered early serotonin signaling either via gestational serotonin reuptake inhibitor (SRI) exposure or genetic variations in the serotonin transporter promoter region (SLC6A4) alters levels of reelin, an important glycoprotein in neurodevelopment, in mothers and their neonates. Serum reelin protein expression was quantified by immunoblot from maternal and neonatal blood collected at delivery from women taking either an SRI during gestation or controls. SRI-exposed mothers had higher levels of one reelin fragment, while SRI-exposed neonates had lower total reelin levels, particularly in females and reelin levels differed with SLC6A4 genotype. Lower neonatal reelin levels predicted less time spent sleeping and more irritability during neonatal behavioral assessment on Day 6 of life. Our results suggest that prenatal SRI exposure and the SLC6A4 genotype influences reelin protein expression in both the mother and newborn and that this may be reflected in neonatal behavior.


Subject(s)
Antidepressive Agents/therapeutic use , Cell Adhesion Molecules, Neuronal/blood , Extracellular Matrix Proteins/blood , Nerve Tissue Proteins/blood , Polymorphism, Single Nucleotide , Prenatal Exposure Delayed Effects/blood , Serine Endopeptidases/blood , Serotonin Plasma Membrane Transport Proteins/genetics , Adult , Antidepressive Agents/pharmacology , Depressive Disorder/drug therapy , Depressive Disorder/genetics , Female , Genotype , Humans , Infant, Newborn , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Reelin Protein , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use
10.
Behav Brain Res ; 237: 333-7, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23047058

ABSTRACT

Accumulating evidence has revealed that dysregulation of the endocannabinoid system could contribute to the development of major depression. Studies carried out post-mortem in depressed suicide victims have revealed increased CB(1) receptor binding site density in the prefrontal cortex (PFC). Accordingly, exposure of rodents to chronic unpredictable stress (CUS) results in phenotypic changes that mirror those of human depression, including increased CB(1) receptor binding site density in the PFC. Our goal in these studies was to examine the effects of CUS on the density of CB(1) receptor binding sites in the rodent medial PFC and to explore the role of this alteration in the behavioral changes invoked by CUS. Rodents exposed to CUS exhibited increased CB(1) receptor maximal binding site density (B(max)) within the ventromedial PFC, but not the dorsomedial PFC. To determine whether this change in the ventromedial PFC is an adaptive response, or alternatively, a consequence of chronic stress that contributes to the adoption of passive coping, we examined whether local CB(1) receptor blockade within the ventromedial PFC following CUS would significantly alter behaviors in the forced swim test (FST). CUS exposure significantly increased passive coping in the FST, and this was further augmented by discrete ventromedial PFC microinfusions of the CB(1) receptor antagonist AM251 prior to swim stress. Moreover, local CB(1) receptor blockade reduced active coping responses in CUS-exposed rats. These findings suggest that the increase in CB(1) receptor B(max) observed in the ventromedial PFC of rodents exposed to CUS maintains proactive coping strategies following chronic stress exposure.


Subject(s)
Prefrontal Cortex/metabolism , Receptor, Cannabinoid, CB1/metabolism , Stress, Psychological/pathology , Up-Regulation , Adaptation, Psychological , Analgesics/pharmacokinetics , Animals , Cues , Cyclohexanols/pharmacokinetics , Disease Models, Animal , Male , Protein Binding , Rats , Rats, Sprague-Dawley , Swimming/psychology , Tritium/pharmacokinetics
11.
J Neuroendocrinol ; 23(11): 1156-65, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21623960

ABSTRACT

Postpartum depression affects 15% of new mothers and previous depressive episodes increase the risk for postpartum depression. Chronic administration of corticosterone (CORT) to dams during the postpartum period causes depressive-like behaviour and has been used as a model of postpartum depression. To better understand the subsequent progress of this model, we examined whether there were persistent effects of CORT treatment during the dam's first postpartum period on maternal care and mood following a subsequent pregnancy. Sprague-Dawley female rats received either sesame oil (control) or CORT (40 mg/kg) injections for 22 days during their first postpartum period. Subsequently, all females were re-mated for a second time and neither group received treatment during the second postpartum period. Maternal care was observed from days 2-8 of each postpartum period and dams were tested in the forced-swim test on days 21 and 22 of the first and days 4 and 21 of the second postpartum period. As expected, the amount of time spent immobile in the forced-swim test was increased in CORT dams at the end of the first postpartum period; however, the amount of time spent immobile was decreased at the end of the second postpartum period relative to oil dams. Furthermore, dams treated with CORT in first postpartum period gave birth to a smaller litter with a larger male/female sex ratio after their second pregnancy. This implies that elevated stress hormone levels during the first postpartum period have a substantial influence on subsequent postpartum behaviour and litter characteristics. Further investigations are necessary to fully understand the effect of parity, experience during first motherhood, and hypothalamic-pituitary-adrenal axis regulation on postpartum depression.


Subject(s)
Behavior, Animal , Corticosterone/blood , Postpartum Period , Pregnancy Outcome , Animals , Female , Male , Pregnancy , Radioimmunoassay , Rats, Sprague-Dawley , Sex Ratio
12.
Neuroscience ; 172: 342-54, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20955769

ABSTRACT

Adult neurogenesis continues throughout life in the mammalian hippocampus and evidence suggests that adult neurogenesis is involved in hippocampus-dependent learning and memory. Numerous studies have demonstrated that spatial learning enhances neurogenesis in the hippocampus but few studies have examined whether enhanced neurogenesis is related to enhanced activation of new neurons in response to spatial learning. Furthermore, the majority of these studies have utilized Sprague-Dawley (SD) rats. However, Long-Evans and Sprague-Dawley rats have been reported to have different learning abilities. In order to determine whether these strains exhibit a similar enhancement of neurogenesis and new neuronal activation in response to spatial learning we tested both strains in a hippocampus-dependent or hippocampus-independent version of the Morris water task (MWT) and then compared levels of neurogenesis and activation of these new cells in the hippocampus. Here we show that despite equivalent performance in the MWT, spatial learning produced a different effect on neurogenesis in each strain. Spatial learning increased cell survival and the number of immature neurons in SD rats compared to cage control and cue-trained rats. In Long-Evans (LE) rats however, spatial learning increased cell survival (BrdU-labeling) but did not increase the number of immature neurons (doublecortin-labeling). Furthermore, we report here an intriguing difference in the activation of new neurons (using the immediate early gene product zif268) in SD versus LE rats. In SD rats we show that spatial learning increases the percentage of doublecortin-labeled cells that are activated during a probe trial. Conversely, in LE rats spatial learning increased the activation of BrdU-labeled but not doublecortin-labeled cells. This interesting difference suggests that different ages or maturational stages of cells are recruited by spatial learning in the two strains. These findings may lead to a better understanding of how and why neurogenesis is regulated by spatial learning.


Subject(s)
Cell Differentiation/physiology , Dentate Gyrus/physiology , Learning/physiology , Neurogenesis/physiology , Neurons/physiology , Space Perception/physiology , Animals , Cell Proliferation , Dentate Gyrus/cytology , Doublecortin Protein , Male , Maze Learning/physiology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurons/cytology , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Species Specificity , Stem Cells/cytology , Stem Cells/physiology
13.
Stress ; 13(4): 301-13, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20536332

ABSTRACT

In adulthood, both alcohol (ethanol) and stress are known to suppress hippocampal neurogenesis in male rats. Similarly, most studies report that prenatal alcohol exposure (PAE) reduces cell proliferation and/or cell survival in the hippocampus of adult males. Furthermore, PAE is known to have marked effects on behavioral and hypothalamic-pituitary-adrenal (HPA) responsiveness to stressors. However, no studies have examined the modulation of adult hippocampal neurogenesis by stress in PAE animals. We hypothesized that, in accordance with previous data, PAE would suppress basal levels of adult hippocampal neurogenesis, and further that stress acting on a sensitized HPA axis would have greater adverse effects on adult hippocampal neurogenesis in PAE than in control rats. Adult male offspring from PAE, pair-fed (PF) control, and ad libitum-fed control (C) groups were subjected to restraint stress (9 days, 1 h/day) or left undisturbed. Rats were then injected with bromodeoxyuridine (BrdU) on day 10, perfused 24 h (proliferation) or 3 weeks (survival) later, and brains processed for BrdU immunohistochemistry. We found that (1) under non-stressed conditions, PAE rats had a small but statistically significant suppressive effect on levels of hippocampal neurogenesis and (2) unexpectedly, repeated restraint stress significantly reduced neurogenesis in C and PF, but not PAE rats. We speculate that the failure of PAE males to mount an appropriate (i.e. suppressive) neurogenic response to stressors, implies reduced plasticity and adaptability or resilience, which could impact negatively on hippocampal structure and function.


Subject(s)
Ethanol/pharmacology , Hippocampus/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/physiopathology , Animals , Corticosterone/metabolism , Female , Hippocampus/drug effects , Hypothalamo-Hypophyseal System/drug effects , Male , Neurons/physiology , Pituitary-Adrenal System/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Restraint, Physical
14.
Neuroscience ; 168(3): 680-90, 2010 Jul 14.
Article in English | MEDLINE | ID: mdl-20406669

ABSTRACT

Adult neurogenesis in the dentate gyrus of the hippocampus is altered with stress exposure and has been implicated in depression. High levels of corticosterone (CORT) suppress neurogenesis in the dentate gyrus of male rats. However both acute and chronic stress do not consistently reduce adult hippocampal neurogenesis in female rats. Therefore, this study was conducted to investigate the effect of different doses of corticosterone on hippocampal neurogenesis in male and female rats. Rats received 21 days of s.c. injections of either oil, 10 or 40 mg/kg CORT. Subjects were perfused 24 h after the last CORT injection and brains were analyzed for cell proliferation (Ki67-labeling) or immature neurons (doublecortin-labeling). Results show that in both males and females high CORT, but not low CORT, reduced both cell proliferation and the density of immature neurons in the dentate gyrus. Furthermore, high CORT males had reduced density in immature neurons in both the ventral and dorsal regions while high CORT females only showed the reduced density of immature neurons in the ventral hippocampus. The high dose of CORT disrupted the estrous cycle of females. Further, the low dose of CORT significantly reduced weight gain and increased basal CORT levels in males but not females, suggesting a greater vulnerability in males with the lower dose of CORT. Thus we find subtle sex differences in the response to chronic CORT on both body weight and on neurogenesis in the dorsal dentate gyrus that may play a role in understanding different vulnerabilities to stress-related neuropsychiatric disorders between the sexes.


Subject(s)
Corticosterone/pharmacology , Dentate Gyrus/drug effects , Animals , Body Weight/drug effects , Cell Count , Cell Proliferation , Cell Survival/drug effects , Corticosterone/blood , Dentate Gyrus/anatomy & histology , Dentate Gyrus/cytology , Doublecortin Protein , Estrus/drug effects , Female , Male , Neurogenesis , Neurons/cytology , Neurons/drug effects , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Sex Factors
15.
J Neuroendocrinol ; 22(4): 248-57, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20136685

ABSTRACT

Motherhood differentially affects learning and memory performance and this effect depends on reproductive experience. In turn, evidence suggests that the effects of oestradiol on learning and memory are mediated through binding to oestrogen receptors in the hippocampus and that this is related to hippocampal neurogenesis. The present study investigated the effect of pregnancy and reproductive experience on ERalpha expression throughout the hippocampus, as well as cell proliferation, new cell survival and cell death (as measured by pyknotic cells) in the granule cell layer of the hippocampus. Three groups of female Sprague-Dawley rats were used: virgin, primigravid and multigravid. All rats were injected with 5-bromo-2-deoxyuridine (BrdU; 200 mg/kg) on the afternoon of impregnation and at matched time-points in virgins. Rats were perfused either during early pregnancy (gestation day 1) or late pregnancy (gestation day 21) after BrdU injection. The results obtained show that, during late pregnancy, females, whether first or second pregnancy, have fewer ERalpha-positive cells in the CA3 region of the dorsal hippocampus than virgin females. In addition during early pregnancy, females have significantly fewer pyknotic cells in the granule cell layer than virgin females. There were no other differences between groups in the number of ERalpha-positive, BrdU-positive or pyknotic cells. Future studies will aim to investigate the mechanisms and consequences of the alteration in ERalpha expression in the hippocampus during late pregnancy, as well as the possible changes in ERbeta expression at this time.


Subject(s)
Cell Proliferation , Estrogen Receptor alpha/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Pregnancy/metabolism , Animals , Apoptosis/physiology , Cell Survival/physiology , Down-Regulation , Female , Gestational Age , Litter Size/physiology , Male , Pregnancy/physiology , Rats , Rats, Sprague-Dawley
16.
J Neuroendocrinol ; 21(3): 155-66, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19076272

ABSTRACT

Oestrogens are known to exert significant structural and functional effects in the hippocampus of adult rodents. The dentate gyrus of the hippocampus retains the ability to produce neurones throughout adulthood and 17beta-oestradiol has been shown to influence hippocampal neurogenesis in adult female rats. The effects of other oestrogens, such as oestrone and 17alpha-oestradiol, on neurogenesis have not been investigated. The present study aimed to investigate the effects of 17beta-oestradiol, oestradiol benzoate, oestrone, and 17alpha-oestradiol on cell proliferation in ovariectomised adult female rats at two different time points. Young ovariectomised female rats were injected with one of the oestrogens at one of three doses. In Experiment 1, rats were exposed to the hormone for 4 h and, in Experiment 2, rats were exposed to the hormone for 30 min prior to 5-bromo-2-deoxyuridine injection to label proliferating cells and their progeny. We found that young ovariectomised females responded with increased cell proliferation to most oestrogens, except oestradiol benzoate, after 30 min of exposure. However, administration of oestrogens for a longer time interval was ineffective at increasing cell proliferation. After 30 min, 17beta-oestradiol and oestrone increased cell proliferation at low (0.3 microg) and high (10 microg) doses, whereas 17alpha-oestradiol increased cell proliferation at medium (1 microg) and high doses. The results of the present study indicate that different oestrogens rapidly increase cell proliferation in a dose-dependent manner, possibly through a nonclassical, nongenomic mechanism. Future experiments should focus on further elucidating the specific pathways utilised by each oestrogen. These results have important therapeutic implications because it may be possible to use 17alpha-oestradiol and lower doses of oestrogens in hormone replacement therapies.


Subject(s)
Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Estrogens/chemistry , Estrogens/pharmacology , Age Factors , Algorithms , Animals , Dentate Gyrus/physiology , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrone/pharmacology , Female , Neurogenesis/drug effects , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Time Factors , Up-Regulation/drug effects
17.
Neuroscience ; 152(4): 888-902, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18353559

ABSTRACT

Estradiol has been shown to have neuroprotective effects, and acute estradiol treatment enhances hippocampal neurogenesis in the female brain. However, little is known about the effects of repeated administration of estradiol on the female brain, or about the effects of estradiol on the male brain. Gonadectomized male and female adult rats were injected with 5-bromo-2-deoxyuridine (BrdU) (200 mg/kg), and then 24 h later were given subcutaneous injections of either estradiol benzoate (33 mug/kg) or vehicle daily for 15 days. On day 16, animals were perfused and the brains processed to examine cells expressing Ki-67 (cell proliferation), BrdU (cell survival), doublecortin (young neuron production), pyknotic morphology (cell death), activated caspase-3 (apoptosis), and Fluoro-Jade B (degenerating neurons) in the dentate gyrus. In female rats, repeated administration of estradiol decreased the survival of new neurons (independent of any effects on initial cell proliferation), slightly increased cell proliferation, and decreased overall cell death in the dentate gyrus. In male rats, repeated administration of estradiol had no significant effect on neurogenesis or cell death. We therefore demonstrate a clear sex difference in the response to estradiol of hippocampal neurogenesis and apoptosis in adult rats, with adult females being more responsive to the effects of estradiol than males.


Subject(s)
Cell Proliferation/drug effects , Estradiol/analogs & derivatives , Hippocampus/cytology , Hippocampus/drug effects , Neurons/drug effects , Sex Characteristics , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Castration/methods , Cell Count , Cell Death/drug effects , Doublecortin Domain Proteins , Doublecortin Protein , Enzyme-Linked Immunosorbent Assay/methods , Estradiol/administration & dosage , Estradiol/blood , Female , Ki-67 Antigen/metabolism , Male , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Rats , Rats, Sprague-Dawley
18.
Neuroscience ; 149(1): 53-67, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17869008

ABSTRACT

Pregnancy and the postpartum period are a time of maximal neural and behavioral plasticity. Recent work has shown that hippocampus-dependent learning and memory performance and hippocampus morphology are affected by motherhood and reproductive experience (number of times pregnant and given birth). Adult neurogenesis in the dentate gyrus of the hippocampus is influenced by steroid hormones such as estradiol and corticosterone, which fluctuate during pregnancy and the postpartum period. Thus, it is possible that hippocampal neurogenesis may be affected by motherhood and reproductive experience. The present study aimed to investigate the role of reproductive experience on hippocampal neurogenesis via cell proliferation and cell survival and to determine whether differences were due to the effect of pregnancy and/or pup-exposure alone. Four groups of female Sprague-Dawley rats were used; multiparous, primiparous, nulliparous, and nulliparous rats exposed to pups. All rats were injected with 5-bromo-2-deoxyuridine (BrdU) (200 mg/kg) approximately 24 h after birth/pup-exposure with age-matched controls. Rats were perfused either 24 h (Expt. 1: Cell proliferation) or 21 days (Expt. 2: Cell survival) after BrdU injection. Results show there is a significant decrease in cell proliferation in the dentate gyrus of primiparous and multiparous rats during the early postpartum period, and a decrease in cell survival in the dentate gyrus during the postpartum in primiparous rats, regardless of pup-exposure, compared with all other groups. In addition, brief pup exposure to nulliparous rats significantly increased cell proliferation and cell death in the dentate gyrus, while 22 days of pup exposure to nulliparous rats (sensitized rats) resulted in increased cell survival and cell death in the dentate gyrus. Collectively these results indicate that reproductive experience significantly affects hippocampal neurogenesis and that these effects are not due to the effect of pregnancy or pup-exposure alone.


Subject(s)
Adult Stem Cells/physiology , Cell Differentiation/physiology , Cell Proliferation , Hippocampus/cytology , Neurons/physiology , Postpartum Period/physiology , Reproduction/physiology , Animals , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Survival/physiology , Estradiol/blood , Estrous Cycle , Female , Male , Maternal Behavior , Pregnancy , Rats
19.
Neuroscience ; 149(2): 273-85, 2007 Oct 26.
Article in English | MEDLINE | ID: mdl-17900815

ABSTRACT

Adult neurogenesis in the hippocampus continues throughout life and may play an important role in hippocampus-dependent learning and memory. Previous research has been equivocal, demonstrating that spatial learning may enhance, decrease or not significantly affect the survival of new neurons. A potential cause of these varying results may be differences in when bromodeoxyuridine (BrdU) was administered relative to spatial training. We examined whether the time elapsed between BrdU administration and spatial learning would alter the survival of the labeled cells. We injected rats with BrdU once on day 0 and then trained in the standard place version of the Morris water task on days 1-5, 6-10 or 11-15 after BrdU injection. We found an enhancement of neurogenesis in the hippocampus only when BrdU was administered 6 days prior to the beginning of spatial training. There was no significant change in hippocampal neurogenesis for groups that started training either 1 or 11 days following BrdU administration. This suggests that a critical period exists in the development of new neurons during which time their survival may be altered by activation of the hippocampus. Furthermore, when dividing rats into poor versus good learners based on overall performance using a median split, only poor place learners and not good place learners exhibit increased hippocampal neurogenesis compared with cue learning, collapsed across time of training. These findings provide further evidence of a link between learning and adult neurogenesis.


Subject(s)
Dentate Gyrus/cytology , Dentate Gyrus/physiology , Hippocampus/physiology , Learning/physiology , Neurons/physiology , Animals , Antimetabolites , Bromodeoxyuridine , Cell Count , Cell Survival/physiology , Cues , DNA/biosynthesis , Data Interpretation, Statistical , Immunohistochemistry , Individuality , Male , Maze Learning/physiology , Psychomotor Performance/physiology , Rats , Rats, Sprague-Dawley
20.
Neuroscience ; 141(4): 1793-800, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16797852

ABSTRACT

This study investigated the involvement of estrogen receptors alpha and beta in estradiol-induced enhancement of hippocampal neurogenesis in the adult female rat. Subtype selective estrogen receptor agonists, propyl-pyrazole triol (estrogen receptor alpha agonist) and diarylpropionitrile (estrogen receptor beta agonist) were examined for each receptor's contribution, individual and cooperative, for estradiol-enhanced hippocampal cell proliferation. Estradiol increases hippocampal cell proliferation within 4 h [Ormerod BK, Lee TT, Galea LA (2003) Estradiol initially enhances but subsequently suppresses (via adrenal steroids) granule cell proliferation in the dentate gyrus of adult female rats. J Neurobiol 55:247-260]. Therefore, animals received s.c. injections of estradiol (10 microg), propyl-pyrazole triol and diarylpropionitrile alone (1.25, 2.5, 5.0 mg/0.1 ml dimethylsulfoxide) or in combination (2.5 mg propyl-pyrazole triol+2.5 mg diarylpropionitrile/0.1 ml dimethylsulfoxide) and 4 h later received an i.p. injection of the cell synthesis marker, bromodeoxyuridine (200 mg/kg). Diarylpropionitrile enhanced cell proliferation at all three administered doses (1.25 mg, P<0.008; 2.5 mg, P<0.003; 5 mg, P<0.005), whereas propyl-pyrazole triol significantly increased cell proliferation (P<0.0002) only at the dose of 2.5 mg. Our results demonstrate both estrogen receptor alpha and estrogen receptor beta are individually involved in estradiol-enhanced cell proliferation. Furthermore both estrogen receptor alpha and estrogen receptor beta mRNA was found co-localized with Ki-67 expression in the hippocampus albeit at low levels, indicating a potential direct influence of each receptor subtype on progenitor cells and their progeny. Dual receptor activation resulted in reduced levels of cell proliferation, supporting previous studies suggesting that estrogen receptor alpha and estrogen receptor beta may modulate each other's activity. Our results also suggest that a component of estrogen receptor-regulated cell proliferation may take place through alternative ligand and/or cell-signaling mechanisms.


Subject(s)
Cell Proliferation/drug effects , Dentate Gyrus/cytology , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Nitriles/pharmacology , Propionates/pharmacology , Pyrazoles/pharmacology , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Count/methods , Dose-Response Relationship, Drug , Doublecortin Domain Proteins , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , In Situ Hybridization/methods , Ki-67 Antigen/metabolism , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Phenols , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...