Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Neuroendocrinol ; 35(1): e13228, 2023 01.
Article in English | MEDLINE | ID: mdl-36690381

ABSTRACT

Hippocampal neuropathology is a recognized feature of the spontaneously hypertensive rat (SHR). The hippocampal alterations associate with cognitive impairment. We have shown that hippocampal abnormalities are reversed by 17ß-estradiol, a steroid binding to intracellular receptors (estrogen receptor α and ß subtypes) or the membrane-located G-protein coupled estradiol receptor. Genistein (GEN) is a neuroprotective phytoestrogen which binds to estrogen receptor ß and G-protein coupled estradiol receptor. Here, we investigated whether GEN neuroprotection extends to SHR. For this purpose, we treated 5-month-old SHR for 2 weeks with 10 mg kg-1 daily s.c injections of GEN. We analyzed the expression of doublecortin+ neuronal progenitors, glial fibrillary acidic protein+ astrocytes and ionized calcium-binding adapter molecule 1+ microglia in the CA1 region and dentate gyrus of the hippocampus using immunocytochemistry, whereas a quantitative real-time polymerase chain reaction was used to measure the expression of pro- and anti-inflammatory factors tumor necrosis factor α, cyclooxygenase-2 and transforming growth factor ß. We also evaluated hippocampal dependent memory using the novel object recognition test. The results showed a decreased number of doublecortin+ neural progenitors in the dentate gyrus of SHR that was reversed with GEN. The number of glial fibrillary acidic protein+ astrocytes in the dentate gyrus and CA1 was increased in SHR but significantly decreased by GEN treatment. Additionally, GEN shifted microglial morphology from the predominantly activated phenotype present in SHR, to the more surveillance phenotype found in normotensive rats. Furthermore, treatment with GEN decreased the mRNA of the pro-inflammatory factors tumor necrosis factor α and cyclooxygenase-2 and increased the mRNA of the anti-inflammatory factor transforming growth factor ß. Discrimination index in the novel object recognition test was decreased in SHR and treatment with GEN increased this parameter. Our results indicate important neuroprotective effects of GEN at the neurochemical and behavioral level in SHR. Our data open an interesting possibility for proposing this phytoestrogen as an alternative therapy in hypertensive encephalopathy.


Subject(s)
Genistein , Phytoestrogens , Rats , Animals , Rats, Inbred SHR , Genistein/pharmacology , Phytoestrogens/pharmacology , Phytoestrogens/metabolism , Glial Fibrillary Acidic Protein/metabolism , Receptors, Estradiol/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cyclooxygenase 2/metabolism , Rats, Inbred WKY , Hippocampus/metabolism , Transforming Growth Factor beta/metabolism , Doublecortin Domain Proteins , RNA, Messenger/metabolism
2.
Cell Mol Neurobiol ; 40(5): 711-723, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31784921

ABSTRACT

It is known that spontaneously hypertensive rats (SHR) present a marked encephalopathy, targeting vulnerable regions such as the hippocampus. Abnormalities of the hippocampus of SHR include decreased neurogenesis in the dentate gyrus (DG), partial loss of neurons in the hilus of the DG, micro and astrogliosis and inflammation. It is also known that 17ß-estradiol (E2) exert neuroprotective effects and prevent hippocampal abnormalities of SHR. The effects of E2 may involve a variety of mechanisms, including intracellular receptors of the ERα and ERß subtypes or membrane-located receptors, such as the G protein-coupled estradiol receptor (GPER). We have now investigated the protective role of GPER in SHR employing its synthetic agonist G1. To accomplish this objective, 5 month-old male SHR received 150 µg/day of G1 during 2 weeks. At the end of this period, we analyzed neuronal progenitors by staining for doublecortin (DCX), and counted the number of glial fibrillary acidic protein (GFAP)-labeled astrocytes and Iba1-stained microglial cells by computerized image analysis. We found that G1 activation of GPER increased DCX+ cells in the DG and reduced GFAP+ astrogliosis and Iba1+ microgliosis in the CA1 region of hippocampus. We also found that the high expression of proinflammatory makers IL1ß and cyclooxygenase 2 (COX2) of SHR was decreased after G1 treatment, which correlated with a change of microglia phenotype from the activated to a resting morphology. Additionally, G1 treatment increased the anti-inflammatory factor TGFß in SHR hippocampus. Altogether, our results suggest that activation of GPER plays a neuroprotective role on the encephalopathy of SHR, an outcome resembling E2 effects but avoiding secondary effects of the natural hormone.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Hippocampus/abnormalities , Hippocampus/pathology , Hypertensive Encephalopathy/metabolism , Inflammation/metabolism , Neurogenesis , Receptors, G-Protein-Coupled/metabolism , Animals , Astrocytes/metabolism , Doublecortin Protein , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Glial Fibrillary Acidic Protein , Hypertensive Encephalopathy/drug therapy , Male , Microglia/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , Rats , Rats, Inbred SHR , Receptors, Estradiol/agonists , Receptors, Estradiol/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics
3.
Cell Mol Neurobiol ; 39(4): 483-492, 2019 May.
Article in English | MEDLINE | ID: mdl-30117098

ABSTRACT

Worldwide, raised blood pressure is estimated to affect 35-40% of the adult population and is a main conditioning factor for cardiovascular diseases and stroke. Animal models of hypertension have provided great advances concerning the pathophysiology of human hypertension, as already shown for the deoxycorticosterone-salt treated rat, the Dahl-salt sensitive rat, the Zucker obese rat and the spontaneously hypertensive rat (SHR). SHR has been widely used to study abnormalities of the brain in chronic hypertension. This review summarises present and past evidence that in the SHR, hypertension causes hippocampal tissue damage which triggers a pro-inflammatory feedforward cascade affecting this vulnerable brain region. The cascade is driven by mineralocorticoid receptor (MR) activation responding to endogenous corticosterone rather than aldosterone. Increased MR expression is a generalised feature of the SHR which seems to support first the rise in blood pressure. Then oxidative stress caused by vasculopathy and hypoxia further increases MR activation in hippocampal neurons and glia cells, activates microglia activation and pro-inflammatory mediators, and down-regulates anti-inflammatory factors. In contrast to MR, involvement of the glucocorticoid receptor (GR) in SHR is less certain. GR showed normal expression levels and blockage with an antagonist failed to reduce blood pressure of SHR. The findings support the concept that MR:GR imbalance caused by vasculopathy causes a switch in MR function towards a proverbial "death" receptor.


Subject(s)
Hypertensive Encephalopathy/metabolism , Nervous System/metabolism , Nervous System/pathology , Receptors, Mineralocorticoid/metabolism , Animals , Disease Models, Animal , Humans , Inflammation/pathology , Receptors, Glucocorticoid/metabolism
4.
Neurochem Res ; 42(8): 2178-2190, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28290135

ABSTRACT

The incorporation of newborn neurons with increased synaptic remodeling and activity-dependent plasticity in the dentate gyrus enhances hippocampal-dependent learning performances. Astrocytes and microglial cells are components of the neurogenic niche and regulate neurogenesis under normal and neurophatological conditions leading to functional consequences for learning and memory. Although cognitive impairments were reported in patients after spinal cord injury (SCI), only few studies have considered remote changes in brain structures which are not related with sensory and motor cortex. Thus, we examined neurogenesis and glial reactivity by stereological assessment in dentate gyrus sub-regions after three different intensities of thoracic spinal cord compression in rats. Sixty days after injury we observed a decrease in the Basso-Bresnahan-Beattie locomotor scale scores, rotarod performance and volume of spare tissue that correlated with the severity of the compression. Regarding the hippocampus, we observed that neurogenesis and hilar neurons were reduced after severe SCI, while only neurogenesis decreased in the moderately injured group. In addition, severe SCI induced reactive microglia and astrogliosis in all dentate gyrus sub-regions. Furthermore, the density of reactive microglia increased in the hilus whereas astrogliosis developed in the molecular layer after moderate SCI. No changes were observed in the mildly injured rats. These results suggest glial response and neurogenesis are associated with injury intensity. Interestingly, hippocampal neurogenesis is more sensitive to SCI than astrocytes or microglia reaction, as moderate injury impairs the generation of new neurons without changing glial response in the subgranular zone.


Subject(s)
Hippocampus/pathology , Neurogenesis/physiology , Neuroglia/physiology , Spinal Cord Injuries/pathology , Animals , Hippocampus/metabolism , Locomotion/physiology , Male , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism
5.
J Steroid Biochem Mol Biol ; 146: 15-25, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24736028

ABSTRACT

Estrogens are neuroprotective factors for brain diseases, including hypertensive encephalopathy. In particular, the hippocampus is highly damaged by high blood pressure, with several hippocampus functions being altered in humans and animal models of hypertension. Working with a genetic model of primary hypertension, the spontaneously hypertensive rat (SHR), we have shown that SHR present decreased dentate gyrus neurogenesis, astrogliosis, low expression of brain derived neurotrophic factor (BDNF), decreased number of neurons in the hilus of the dentate gyrus, increased basal levels of the estrogen-synthesizing enzyme aromatase, and atrophic dendritic arbor with low spine density in the CA1 region compared to normotensive Wistar Kyoto (WKY) ratsl. Changes also occur in the hypothalamus of SHR, with increased expression of the hypertensinogenic peptide arginine vasopressin (AVP) and its V1b receptor. Following chronic estradiol treatment, SHR show decreased blood pressure, enhanced hippocampus neurogenesis, decreased the reactive astrogliosis, increased BDNF mRNA and protein expression in the dentate gyrus, increased neuronal number in the hilus of the dentate gyrus, further increased the hyperexpression of aromatase and replaced spine number with remodeling of the dendritic arbor of the CA1 region. We have detected by qPCR the estradiol receptors ERα and ERß in hippocampus from both SHR and WKY rats, suggesting direct effects of estradiol on brain cells. We hypothesize that a combination of exogenously given estrogens plus those locally synthesized by estradiol-stimulated aromatase may better alleviate the hippocampal and hypothalamic encephalopathy of SHR. This article is part of a Special Issue entitled "Sex steroids and brain disorders".


Subject(s)
Estradiol/pharmacology , Estrogens/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Hypertensive Encephalopathy/metabolism , Neuroprotective Agents/pharmacology , Animals , Aromatase/metabolism , Blood Pressure/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Dendrites/drug effects , Dendrites/metabolism , Estradiol/metabolism , Estrogen Receptor alpha , Estrogen Receptor beta , Humans , Rats, Inbred SHR , Rats, Inbred WKY
6.
Exp Neurol ; 247: 158-64, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23628746

ABSTRACT

Increased neuronal vulnerability has been described in the brain of spontaneously hypertensive rats (SHR), models of primary hypertension. Previous data indicate that estradiol treatment corrects several dysfunctions of the hippocampus and hypothalamus of SHR. Considering this evidence we analyzed the dendritic arborization and spine density of the CA1 subfield in SHR and Wistar-Kyoto (WKY) normotensive rats with and without estradiol treatment. Five month old male SHR and WKY rats received single estradiol or cholesterol pellets (sham treatment) for 2 weeks. A substantial rise of circulating estradiol (>25 fold) and testicular atrophy was present in all estradiol-receiving rats. In both SHR and WKY rats, estradiol decreased blood pressure by ~20 mm Hg; however, a moderate hypertension persisted in SHR (164 mm Hg). Using a modified Golgi impregnation technique, apical and basal dendrites of the CA1 subfield were subjected to Sholl analysis. Spine density was also statistically analyzed. Apical dendritic length was significantly lower in SHR compared to WKY rats (p<0.01), whereas estradiol treatment increased dendritic length in the SHR group only (SHR vs SHR+estradiol; p<0.01). Apical dendritic length plotted against the shell distances 20-100, 120-200 and 220-300 µm, revealed that changes were more pronounced in the range 120-200 µm between SHR vs. WKY rats (p<0.05) and SHR vs. SHR+estradiol (p<0.05). Instead, basal dendrites were not significantly modified by hypertension or steroid treatment. Spine density of apical dendrites was lower in SHR than WKY (p<0.05) and was up-regulated in the SHR+estradiol group compared to the SHR group (p<0.001). Similar changes were obtained for basal dendritic spines. These data suggest that changes of neuronal processes in SHR are plastic events restorable by estradiol treatment. In conjunction with previous results, the present data reveal new targets of estradiol neuroprotection in the brain of hypertensive rats.


Subject(s)
CA1 Region, Hippocampal/pathology , Dendrites/ultrastructure , Dendritic Spines/drug effects , Estradiol/pharmacology , Hypertension/pathology , Neurons/cytology , Analysis of Variance , Animals , Atrophy/chemically induced , Blood Pressure/drug effects , Dendrites/drug effects , Disease Models, Animal , Estradiol/blood , Hypertension/drug therapy , Male , Neurons/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Silver Staining , Testis/drug effects
7.
Horm Mol Biol Clin Investig ; 4(2): 549-57, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-25961231

ABSTRACT

Estrogen neuroprotection has been shown in pathological conditions damaging the hippocampus, such as trauma, aging, neurodegeneration, excitotoxicity, oxidative stress, hypoglycemia, amyloid-ß peptide exposure and ischemia. Hypertensive encephalopathy also targets the hippocampus; therefore, hypertension seems an appropriate circumstance to evaluate steroid neuroprotection. Two experimental models of hypertension, spontaneously hypertensive rats (SHR) and deoxycorticosterone (DOCA)-salt hypertensive rats, develop hippocampal abnormalities, which include decreased neurogenesis in the dentate gyrus, astrogliosis, low expression of brain-derived neurotrophic factor (BDNF) and decreased number of neurons in the hilar region, with respect of their normotensive strains Wistar Kyoto (WKY) and Sprague-Dawley rats. After estradiol was given for 2 weeks to SHR and DOCA-treated rats, both hypertensive models normalized their faulty hippocampal parameters. Thus, estradiol treatment positively modulated neurogenesis in the dentate gyrus of the hippocampus, according to bromodeoxyuridine incorporation and doublecortin immunocytochemistry, decreased reactive astrogliosis, increased BDNF mRNA and protein expression in the dentate gyrus and increased neuronal number in the hilar region of the dentate gyrus. A role of local estrogen biosynthesis is suggested in SHR, because basal aromatase mRNA in the hippocampus and immunoreactive aromatase protein in cell processes of the dentate gyrus were highly expressed in these rats. Estradiol further stimulated aromatase-related parameters in SHR but not in WKY. These observations strongly support that a combination of exogenous estrogens to those locally synthesized might better alleviate hypertensive encephalopathy. These studies broaden estrogen neuroprotective functions to the hippocampus of hypertensive rat models.

8.
Exp Gerontol ; 44(1-2): 34-40, 2009.
Article in English | MEDLINE | ID: mdl-18445514

ABSTRACT

Neuroactive steroids are secretory products of peripheral endocrine glands that modulate a variety of brain functions. A close relationship between neuroactive steroid structure and function becomes most evident under pathological circumstances. On one side, overproduction of glucocorticoid and mineralocorticoid neuroactive steroids may be detrimental to the hippocampus, which is enriched in glucocorticoid receptors (GR) and mineralocorticoid receptors (MR). Thus, a dysfunction of the adrenocortical system in aging and age-associated diseases (diabetes, hypertension) is able to cause hippocampal damage. Whereas aging and uncontrolled diabetes show a predominant GR overdrive, a MR overdrive characterizes hypertensive animals. Some abnormalities commonly found in the hippocampus of aging, diabetic and hypertensive animals include decreased neurogenesis, astrogliosis and neuronal loss in the hilus of the dentate gyrus (DG). On the other side, and in contrast to adrenal gland-derived steroids, estrogens qualify as hippocampal neuroprotectants. Given to middle-age mice, estrogens stimulated proliferation and differentiation of newborn cells in the DG, decreased astrogliosis and increased hilar neuronal number. Similar estrogen effects were obtained in mice with streptozotocin-induced diabetes and in spontaneously hypertensive rats (SHR). The results suggest that in aging and age-associated diseases, adrenocortical steroid overdrive sensitizes the hippocampus to the pathological milieu imposed by a pre-existing degeneration or illness. In this setting, estradiol neuroprotection rescues hippocampal parameters previously altered by the pathological environment.


Subject(s)
Aging/physiology , Estradiol/pharmacology , Hippocampus/metabolism , Neuroprotective Agents/pharmacology , Adrenal Cortex/metabolism , Aged , Animals , Diabetes Mellitus/metabolism , Estradiol/metabolism , Glucocorticoids/metabolism , Humans , Hypertension/metabolism , Middle Aged , Mineralocorticoids/metabolism , Neuroprotective Agents/metabolism , Rats , Rats, Inbred Strains
9.
Psychoneuroendocrinology ; 33(3): 270-81, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18164826

ABSTRACT

Abnormalities of hippocampus and hypothalamus are commonly observed in rats with genetic (SHR) or mineralocorticoid/salt-induced hypertension. In the hippocampus, changes include decreased cell proliferation in the dentate gyrus (DG), astrogliosis and decreased neuronal density in the hilus, whereas in the hypothalamus expression of arginine vasopressin (AVP) is markedly elevated. Here, we report that estradiol treatment overturns these abnormalities. We used 16-week-old male SHR with blood pressure (BP) approximately 190 mmHg and their normotensive Wistar-Kyoto (WKY) controls, and male Sprague-Dawley rats made hypertensive by administration of 10mg deoxycorticosterone acetate (DOCA) every other day plus 1% NaCl as drinking fluid for 4 weeks (BP approximately 160 mmHg). Controls received oil vehicle plus 1% NaCl only. Half of the animals in each group were implanted s.c. with a single estradiol benzoate pellet weighing 14 mg for 2 weeks. Estradiol-treated SHR and DOCA-salt rats showed, in comparison to their respective steroid-free groups: (a) enhanced proliferation in the DG measured by bromodeoxyuridine incorporation; (b) decreased number of glial fibrillary acidic protein (GFAP) immunopositive astrocytes; (c) increased density of neurons in the hilus of the DG, and (d) decreased hypothalamic AVP mRNA expression. These results indicate that neuronal and glial alterations of hypertensive models are plastic events reversible by steroid treatment. The estradiol protective effects may be of pharmacological interest to attenuate the consequences of hypertensive encephalopathy.


Subject(s)
Brain/pathology , Estradiol/pharmacology , Hypertension/pathology , Mineralocorticoids , Neuroprotective Agents , Animals , Arginine Vasopressin/biosynthesis , Bromodeoxyuridine/pharmacology , Cell Proliferation/drug effects , Dentate Gyrus/pathology , Desoxycorticosterone , Glial Fibrillary Acidic Protein/metabolism , Hypertension/chemically induced , Hypertension/genetics , Immunohistochemistry , In Situ Hybridization , Male , Neurons/pathology , Neurons/ultrastructure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley
10.
Psychoneuroendocrinology ; 32(5): 480-92, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17459595

ABSTRACT

During aging the hippocampus experiences structural, molecular, and functional alterations. Protection from age-related disorders is provided by several factors, including estrogens. Since aging defects start at middle age, we studied if 17 beta-estradiol (E(2)) protected the hippocampus at this age period. Middle age (10-12 month old) male C57Bl/6 mice were implanted sc with E(2) (15 microg) or cholesterol pellets. Ten days afterwards they received bromodeoxyuridine (BrdU) 4 and 2h before killing to study cell proliferation in the dentate gyrus (DG). A pronounced depletion of BrdU+cells in the DG was found in cholesterol-treated middle age mice, accompanied by astrocytosis, and by neuronal loss in the hilus. Middle age mice receiving E(2) showed increased number of BrdU+cells while the other parameters were remarkably attenuated. When steroid treatment was prolonged for 2 months to study migration of cells in the granular layer of the DG, cell migration was unaffected by E(2). However, E(2)-treated middle age mice presented higher cell density and increased staining for doublecortin, a marker for differentiating neurons. Thus, from the three basic steps of adult neurogenesis (proliferation, migration, and differentiation), E(2) stimulated progenitor proliferation - even after long exposure to E(2) studied by Ki67 immunocytochemistry - and differentiation towards a neuronal lineage. This result, in conjunction with recovery from other aging indicators as increased deposits of the aging pigment lipofuscin in DG cells, loss of hilar neurons and astrocytosis supports a wide range protection of hippocampal function of middle age mice by estrogenic hormones.


Subject(s)
Cell Differentiation/physiology , Estradiol/metabolism , Hippocampus/metabolism , Neuroglia/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Aging/physiology , Animals , Cell Death/physiology , Hippocampus/cytology , Immunohistochemistry , Male , Mice , Neuroglia/cytology , Neurons/cytology , Sex Factors , Stem Cells/cytology , Stem Cells/metabolism
11.
Front Horm Res ; 35: 157-168, 2006.
Article in English | MEDLINE | ID: mdl-16809931

ABSTRACT

The function of the HPA axis is subject to regulation by many factors, which achieve relevance under normal and pathological conditions. In the case of aging, this period of life is associated with disturbances of the HPA axis and signs of hippocampal vulnerability. We examined 20-month-old male rats, in which abnormalities of the HPA axis included altered response to stress, reduced effectiveness of the steroid negative feedback and low expression of hippocampal glucocorticoid receptors (GR). Estrogen treatment of aging rats normalized the response to stress, restored the dexamethasone inhibition of the stress response and increased GR density in defined hippocampal areas. Although estrogens could influence the hippocampus of aging animals directly, their effects could be also mediated by estrogen-sensitive forebrain cholinergic neurons projecting to the hippocampus. Additionally, estrogens normalized the deficient granule cell proliferation that aging mice present in the dentate gyrus, and attenuated several markers of hippocampal aging, such as astrocytosis, high lipofucsin content and neuronal loss in the hilus of the dentate gyrus. These effects may be important for the regulation of the HPA axis, in the context that hippocampal function as a whole was normalized by estrogen action. Therefore, estrogens are powerful neuroprotectants in cases of hippocampal dysfunction, and as part of this effect, they contribute to stabilize the function of the HPA axis.


Subject(s)
Adrenal Glands/physiology , Aging , Estrogens/pharmacology , Hypothalamo-Hypophyseal System/physiology , Neurosecretory Systems/physiology , Pituitary-Adrenal System/physiology , Animals , Cholinergic Fibers/metabolism , Feedback, Physiological/physiology , Hippocampus/drug effects , Humans , Prosencephalon/metabolism , Rats , Steroids/physiology
12.
Neuroendocrinology ; 80(2): 100-10, 2004.
Article in English | MEDLINE | ID: mdl-15475662

ABSTRACT

Mineralocorticoid effects in the brain include the control of cardiovascular functions, induction of salt appetite, interaction with the vasoactive neuropeptides arginine vasopressin (AVP) and angiotensin II and development or aggravation of hypertension. In this regard, mineralocorticoids may play a pathogenic role in rats with a genetic form of hypertension (spontaneously hypertensive rats, SHR). Our objective was to compare the response of the hypothalamic vasopressinergic system to mineralocorticoid administration in SHR and control Wistar-Kyoto (WKY) rats. Sixteen-week-old male SHR showing a systolic blood pressure of 190 +/- 5 mm Hg and normotensive WKY rats (130 +/- 5 mm Hg) were treated subcutaneously with oil vehicle or a single 10-mg dose of deoxycorticosterone acetate (DOCA). After 2 h, rats were sacrificed and brains prepared for immunocytochemistry of Fos and vasopressin V1a receptor (V1aR) and for non-isotopic in situ hybridization of AVP mRNA. In the basal state, SHR demonstrated a higher number of AVP mRNA- and V1aR-immunopositive cells in the magnocellular division of the paraventricular hypothalamic nucleus (PVN) than WKY rats. After DOCA injection, SHR responded with a significant increase in both parameters with respect to vehicle-injected SHR. In WKY rats, DOCA was without effect on AVP mRNA although it increased the number of V1aR-positive cells. Changes in the number of Fos-positive nuclei were measured in the PVN, median preoptic nucleus (MnPO) and organum vasculosum of the lamina terminalis (OVLT), a circumventricular region showing anatomical connections with the PVN. In vehicle-injected rats, the PVN of SHR showed a higher number of Fos-positive nuclei than in WKY rats, whereas after DOCA treatment, a significant increment occurred in the OVLT but not in the PVN or MnPO of the SHR group only. These data suggest that the enhanced response of the vasopressinergic system to mineralocorticoids may contribute to the abnormal blood pressure of SHR.


Subject(s)
Hypertension/physiopathology , Hypothalamus/drug effects , Mineralocorticoids/pharmacology , Vasopressins/drug effects , Animals , Arginine Vasopressin/drug effects , Arginine Vasopressin/metabolism , Desoxycorticosterone/pharmacology , Disease Models, Animal , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Oncogene Proteins v-fos/drug effects , Oncogene Proteins v-fos/metabolism , RNA, Messenger , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Vasopressin/drug effects , Receptors, Vasopressin/metabolism , Vasopressins/metabolism
13.
Neuro Endocrinol Lett ; 23(4): 321-4, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12195234

ABSTRACT

OBJECTIVES: The effects of dehydroepiandrosterone (DHEA) on galanin (GAL) and prolactin (PRL) mRNA expression in the anterior pituitary of Fischer 344 rats were studied, taking in consideration that: (1) DHEA is an androgen with estrogenic activity on pituitary lactotrophs; (2) estrogens induce prolactinomas in Fischer 344 rats; and (3) GAL has been considered the main mediator of estrogen-induced lactotroph proliferation. DESIGN: Female rats were ovariectomized and used as controls or treated during 2 weeks with DHEA (500 mg/kg/day or 5 mg/kg/day or 50 mg/kg/day) or estradiol (E2, 50 microg/kg/day), as a positive control for pituitary growth and GAL induction. GAL and PRL mRNA expression were studied by in situ hybridization. RESULTS: Both DHEA and E2 induced PRL mRNA synthesis. However, DHEA neither produced pituitary enlargement nor GAL induction, in contrast to E2. CONCLUSIONS: Our results shows that GAL is not involved in the estrogenic activity of DHEA on pituitary lactotrophs, and suggest that DHEA effects are exerted directly on the PRL gene or through another mechanism(s) not related to GAL.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dehydroepiandrosterone/pharmacology , Galanin/genetics , Pituitary Gland, Anterior/physiology , Prolactin/genetics , Animals , Carcinogenicity Tests , Estradiol/pharmacology , Female , Gene Expression/drug effects , Pituitary Gland, Anterior/drug effects , Prolactin/blood , RNA, Messenger/analysis , Rats , Rats, Inbred F344
14.
Neurosci Lett ; 329(3): 344-8, 2002 Sep 06.
Article in English | MEDLINE | ID: mdl-12183045

ABSTRACT

Mineralocorticoids (MC) play an important role in development of salt appetite. Part of this effect involves the hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei, in which MC treatment increases arginine vasopressin (AVP) synthesis and release. Since the AVP system is also modulated by nitric oxide (NO), we studied if deoxycorticosterone acetate (DOCA) treatment changed the number of nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) active neurons and neuronal NO synthase (nNOS)-immunoreactive (IR) cells in the PVN and SON. After four injections of DOCA (10 mg/rat per day), rats developed a salt appetite and increased NADPH-d active and nNOS-IR neurons in both nuclei. A single DOCA injection did not change salt consumption or nNOS-IR cells, but increased the number of NADPH-d positive neurons in the PVN only. Therefore, while acute MC treatment stimulated the activity of pre-existing enzyme, chronic steroid treatment recruited additional neurons showing nNOS immunoreactivity/NADPH-d activity. These data suggest a role for NO produced in the PVN and SON in DOCA stimulatory effects on AVP mRNA and salt appetite.


Subject(s)
Desoxycorticosterone/pharmacology , Hypothalamus, Anterior/enzymology , NADPH Dehydrogenase/analysis , Nitric Oxide Synthase/analysis , Paraventricular Hypothalamic Nucleus/enzymology , Animals , Hypothalamus, Anterior/cytology , Hypothalamus, Anterior/drug effects , Immunohistochemistry , Male , NADPH Dehydrogenase/immunology , Neurons/enzymology , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase Type I , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Sprague-Dawley , Sodium Chloride, Dietary/pharmacology
15.
Neurobiol Dis ; 11(3): 457-68, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12586554

ABSTRACT

Motor neuron degeneration characterizes the spinal cord of patients with amyotrophic lateral sclerosis and the Wobbler mouse mutant. Considering that progesterone (PROG) provides neuroprotection in experimental ischemia and injury, its potential role in neurodegeneration was studied in the murine model. Two-month-old symptomatic Wobbler mice were left untreated or received sc a 20-mg PROG implant for 15 days. Both light and electron microscopy of Wobbler mice spinal cord showed severely affected motor neurons with profuse cytoplasmic vacuolation of the endoplasmic reticulum and/or Golgi apparatus and ruptured mitochondria with damaged cristae, a profile indicative of a type II cytoplasmic form of cell death. In contrast to untreated mice, neuropathology was less severe in Wobbler mice receiving PROG; including a reduction of vacuolation and of the number of vacuolated cells and better conservation of the mitochondrial ultrastructure. In biochemical studies, we determined the mRNA for the alpha3 subunit of Na,K-ATPase, a neuronal enzyme controlling ion fluxes, neurotransmission, membrane potential, and nutrient uptake. In untreated Wobbler mice, mRNA levels in motor neurons were reduced by half compared to controls, whereas PROG treatment of Wobbler mice restored the expression of alpha3 subunit Na,K-ATPase mRNA. Therefore, PROG was able to rescue motor neurons from degeneration, based on recovery of histopathological abnormalities and of mRNA levels of the sodium pump. However, because the gene mutation in Wobbler mice is still unknown, further studies are needed to unveil the action of PROG and the mechanism of neuronal death in this genetic model of neurodegeneration.


Subject(s)
Motor Neuron Disease/metabolism , Motor Neurons/metabolism , Neuroprotective Agents/metabolism , Progesterone/metabolism , Spinal Cord Diseases/metabolism , Animals , In Situ Hybridization , Mice , Mice, Neurologic Mutants , Microscopy, Electron , Motor Neuron Disease/drug therapy , Motor Neurons/enzymology , Motor Neurons/ultrastructure , Neuroprotective Agents/administration & dosage , Progesterone/administration & dosage , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Sodium-Potassium-Exchanging ATPase/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Spinal Cord Diseases/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...