Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
J Neuroendocrinol ; 29(7)2017 07.
Article En | MEDLINE | ID: mdl-28523794

Damage observed in the hippocampus of the adult spontaneously hypertensive rat (SHR) resembles the neuropathology of mineralocorticoid-induced hypertension, supporting a similar endocrine dysfunction in both entities. In the present study, we tested the hypothesis that increased expression of the hippocampal mineralocorticoid receptor (MR) in SHR animals is associated with a prevalent expression of pro-inflammatory over anti-inflammatory factors. Accordingly, in the hippocampus, we measured mRNA expression and immunoreactivity of the MR and glucocorticoid receptor (GR) using a quantitative polymerase chain reaction and histochemistry. We also measured serum-glucocorticoid-activated kinase 1 (Sgk1 mRNA), the number and phenotype of Iba1+ microglia, as well as mRNA expression levels of the pro-inflammatory factors cyclo-oxygenase 2 (Cox2), Nlrp3 inflammasome and tumour necrosis factor α (Tnfα). Expression of anti-inflammatory transforming growth factor (Tgf)ß mRNA and the NADPH-diaphorase activity of nitric oxide synthase (NOS) were also determined. The results showed that, in the hippocampus of SHR rats, expression of MR and the number of immunoreactive MR/GR co-expressing cells were increased compared to Wistar-Kyoto control animals. Expression of Sgk1, Cox2, Nlrp3 and the number of ramified glia cells positive for Iba1+ were also increased, whereas Tgfß mRNA expression and the NADPH-diaphorase activity of NOS were decreased. We propose that, in the SHR hippocampus, increased MR expression causes a bias towards a pro-inflammatory phenotype characteristic for hypertensive encephalopathy.


Hippocampus/metabolism , Inflammation/metabolism , Neurons/metabolism , Receptors, Mineralocorticoid/metabolism , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Male , Microglia/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
2.
J Neuroendocrinol ; 28(10)2016 10.
Article En | MEDLINE | ID: mdl-27517478

Spontaneously hypertensive rats (SHR) show pronounced hippocampus alterations, including low brain-derived neurotrophic factor (BDNF) expression, reduced neurogenesis, astrogliosis and increased aromatase expression. These changes are reverted by treatment with 17ß-oestradiol. To determine which oestradiol receptor (ER) type is involved in these neuroprotective effects, we used agonists of the ERα [propylpyrazole triol (PPT)] and the ERß [diarylpropionitrite (DPN)] given over 2 weeks to 4-month-old male SHR. Wistar Kyoto normotensive rats served as controls. Using immunocytochemistry, we determined glial fibrillary protein (GFAP)+ astrocytes in the CA1, CA3 and hilus of the dentate gyrus of the hippocampus, aromatase immunostaining in the hilus, and doublecortin (DCX)+ neuronal progenitors in the inner granular zone of the dentate gyrus. Brain-derived neurotrophic factor mRNA was also measured in the hippocampus by the quantitative polymerase chain reaction. In SHR, PPT had no effect on blood pressure, decreased astrogliosis, slightly increased BDNF mRNA, had no effect on the number of DCX+ progenitors, and increased aromatase staining. Treatment with DPN decreased blood pressure, decreased astrogliosis, increased BDNF mRNA and DCX+ progenitors, and did not modify aromatase staining. We hypothesise that, although both receptor types may participate in the previously reported beneficial effects of 17ß-oestradiol in SHR, receptor activation with DPN may preferentially facilitate BDNF mRNA expression and neurogenesis. The results of the present study may help in the design of ER-based neuroprotection for the encephalopathy of hypertension.


Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Hippocampus/drug effects , Hippocampus/metabolism , Nitriles/administration & dosage , Phenols/administration & dosage , Propionates/administration & dosage , Pyrazoles/administration & dosage , Animals , Aromatase/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Blood Pressure , Brain-Derived Neurotrophic Factor/metabolism , Doublecortin Protein , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Gliosis , Male , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Organ Size , Pituitary Gland/anatomy & histology , Pituitary Gland/drug effects , RNA, Messenger/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Testis/anatomy & histology , Testis/drug effects
3.
Neuroscience ; 280: 243-53, 2014 Nov 07.
Article En | MEDLINE | ID: mdl-25242645

Previous work has shown a reduction of apical dendritic length and spine density in neurons from the CA1 hippocampus subfield of spontaneously hypertensive rats (SHRs). These abnormalities are prevented by treatment for 2 weeks with 17ß-estradiol. In view of the fact that diabetes and hypertension are comorbid diseases, we have now studied the effect of Streptozotocin-induced diabetes on the dendritic tree and spines of CA1 hippocampus neurons, and also compared the regulation of these parameters by 17ß-estradiol in diabetic and normoglycemic SHR. Twenty-week-old male SHR received i.v. 40-mg/kg Streptozotocin or vehicle and studied 1 month afterward. A group of normoglycemic and hyperglycemic SHR also received s.c. a single 17ß-estradiol pellet or vehicle for 2weeks. Hippocampus sections were impregnated with silver nitrate following a modified Golgi's method and the arbor of CA1 pyramidal neurons analyzed by Sholl's method. 17ß-Estradiol treatment of normoglycemic SHR reversed the reduced length of apical dendrites, the low spine density and additionally decreased blood pressure (BP). Diabetic SHR showed increased length of apical and basal dendrites but reduced spine density compared to normoglycemic SHR. Diabetes also decreased BP of SHR. Treatment with 17ß-estradiol of diabetic SHR enhanced dendritic length, increased dendritic spine density and further decreased BP. Thus, changes of cytoarchitecture of CA1 neurons due to 17ß-estradiol treatment of normoglycemic SHR persisted after diabetes induction. A decrease of BP may also contribute to the central effects of 17ß-estradiol in SHR diabetic rats.


CA1 Region, Hippocampal/drug effects , Dendrites/drug effects , Diabetes Mellitus, Experimental/drug therapy , Estradiol/pharmacology , Neuroprotective Agents/pharmacology , Pyramidal Cells/drug effects , Animals , Blood Pressure/drug effects , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Dendrites/pathology , Dendrites/physiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Hypertension/drug therapy , Hypertension/pathology , Hypertension/physiopathology , Male , Photomicrography , Pyramidal Cells/pathology , Pyramidal Cells/physiopathology , Rats, Inbred SHR
4.
J Neuroendocrinol ; 26(5): 310-20, 2014 May.
Article En | MEDLINE | ID: mdl-24730417

17ß-oestradiol is a powerful neuroprotective factor for the brain abnormalities of spontaneously hypertensive rats (SHR). 17α-Oestradiol, a nonfeminising isomer showing low affinity for oestrogen receptors, is also endowed with neuroprotective effects in vivo and in vitro. We therefore investigated whether treatment with 17α-oestradiol prevented pathological changes of the hippocampus and hypothalamus of SHR. We used 20-week-old male SHR with a blood pressure of approximately 170 mmHg receiving s.c. a single 800 µg pellet of 17α-oestradiol dissolved in cholesterol or vehicle only for 2 weeks Normotensive Wistar-Kyoto (WKY) rats were used as controls. 17α-Oestradiol did not modify blood pressure, serum prolactin, 17ß-oestradiol levels or the weight of the testis and pituitary of SHR. In the brain, we analysed steroid effects on hippocampus Ki67+ proliferating cells, doublecortin (DCX) positive neuroblasts, glial fibrillary acidic protein (GFAP)+ astrocyte density, aromatase immunostaining and brain-derived neurotrophic factor (BDNF) mRNA. In the hypothalamus, we determined arginine vasopressin (AVP) mRNA. Treatment of SHR with 17α-oestradiol enhanced the number of Ki67+ in the subgranular zone and DCX+ cells in the inner granule cell layer of the dentate gyrus, increased BDNF mRNA in the CA1 region and gyrus dentatus, decreased GFAP+ astrogliosis in the CA1 subfield, and decreased hypothalamic AVP mRNA. Aromatase expression was unmodified. By contrast to SHR, normotensive WKY rats were unresponsive to 17α-oestradiol. These data indicate a role for 17α-oestradiol as a protective factor for the treatment of hypertensive encephalopathy. Furthermore, 17α-oestradiol is weakly oestrogenic in the periphery and can be used in males.


Brain/drug effects , Estradiol/pharmacology , Neuroprotective Agents/pharmacology , Animals , Arginine Vasopressin/metabolism , Blood Pressure/drug effects , Brain Chemistry/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Doublecortin Protein , Gliosis/pathology , Male , Neurogenesis/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY
5.
Mini Rev Med Chem ; 12(11): 1081-9, 2012 Oct.
Article En | MEDLINE | ID: mdl-22827218

Besides their effects on reproduction, estrogens exert neuroprotective effects for brain diseases. Thus, estrogens ameliorate the negative aspects of aging and age-associated diseases in the nervous system, including hypertension. Within the brain, the hippocampus is sensitive to the effects of hypertension, as exemplified in a genetic model, the spontaneously hypertensive rat (SHR). In the dentate gyrus of the hippocampus, SHR present decreased neurogenesis, astrogliosis, low expression of brain derived neurotrophic factor (BDNF), decreased number of neurons in the hilus and increased basal levels of the estrogen-synthesizing enzyme aromatase, with respect to the Wistar Kyoto (WKY) normotensive strain. In the hypothalamus, SHR show increased expression of the hypertensinogenic peptide arginine vasopressin (AVP) and its V1b receptor. From the therapeutic point of view, it was highly rewarding that estradiol treatment decreased blood pressure and attenuated brain abnormalities of SHR, rendering hypertension a suitable model to test estrogen neuroprotection. When estradiol treatment was given for 2 weeks, SHR normalized their faulty brain parameters. This was shown by the enhancement of neurogenesis in the dentate gyrus, according to increased bromodeoxyuridine incorporation and doublecortin labeling, decreased reactive astrogliosis, increased BDNF mRNA and protein expression in the dentate gyrus, increased neuronal number in the hilus of the dentate gyrus and a further hyperexpression of aromatase. The presence of estradiol receptors in hippocampus and hypothalamus suggests the possibility of direct effects of estradiol on brain cells. Successful neuroprotection produced by estradiol in hypertensive rats should encourage the treatment with non-feminizing estrogens and estrogen receptor modulators for age-associated diseases.


Estradiol/metabolism , Estradiol/therapeutic use , Hypertensive Encephalopathy/drug therapy , Hypertensive Encephalopathy/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Animals , Blood Pressure/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Doublecortin Protein , Estradiol/pharmacology , Humans , Hypertensive Encephalopathy/pathology , Hypertensive Encephalopathy/physiopathology , Neuroprotective Agents/pharmacology
6.
J Neuroendocrinol ; 24(9): 1249-58, 2012 Sep.
Article En | MEDLINE | ID: mdl-22564091

The mineralocorticoid receptor (MR) has been considered as both neuroprotective and damaging to the function of the central nervous system. MR may be also involved in central regulation of blood pressure. In the present study, we compared the expression of MR and the glucocorticoid receptor (GR) in the hippocampus and hypothalamus of 16-week-old spontaneously hypertensive rats (SHR) and normotensive control Wistar Kyoto (WKY) rats. In the hippocampus, MR expression was studied by in situ hybridization (ISH), quantitative polymerase chain reaction (PCR) and immunohistochemistry, whereas GR expression was analysed using the latter two procedures. Hypertensive animals showed an increased expression of MR mRNA in the whole hippocampus according to qPCR data and also in CA3 by ISH. Immunocytochemical staining for MR of the dorsal hippocampus, however, did not reveal differences between SHR and WKY rats. SHR showed elevated hypothalamic MR mRNA by qPCR, as well as an increased number of MR immunopositive cells in the magnocellular paraventricular region, compared to WKY rats. By contrast, expression levels of GR mRNA or protein in the hippocampus and hypothalamus of SHR were similar to those of WKY rats. Furthermore, we investigated the role of MR in the hypertensive rats by i.c.v. injection of the MR antagonist RU-2831. This compound produced a significant drop in blood pressure for SHR. In conclusion, MR expression is increased in the hippocampus and hypothalamus of SHR. We suggest that pathological MR overdrive may take responsibility for up-regulation of blood pressure and the encephalopathy of hypertension.


Hippocampus/metabolism , Hypothalamus/metabolism , Rats, Inbred SHR/metabolism , Receptors, Mineralocorticoid/biosynthesis , Animals , Blood Pressure/drug effects , Mineralocorticoid Receptor Antagonists/pharmacology , Rats , Rats, Inbred WKY , Receptors, Glucocorticoid/biosynthesis , Spironolactone/analogs & derivatives , Spironolactone/pharmacology
7.
Neuroscience ; 174: 151-9, 2011 Feb 03.
Article En | MEDLINE | ID: mdl-21115103

There is high incidence of hippocampal abnormalities in spontaneously hypertensive rats (SHR), including decreased neurogenesis in the dentate gyrus, astrogliosis, low expression of brain derived neurotrophic factor and decreased neuronal density in the hilar region, respect of normotensive Wistar Kyoto rats (WKY). Estradiol treatment given for 2 weeks normalized the faulty hippocampal parameters of SHR, without having effects on WKY rats. The present work studied the potential role of local estrogen biosynthesis in the hippocampus of SHR and WKY, by measuring the expression of aromatase, the key enzyme responsible for estrogen biosynthesis and involved in neuroprotection. We used 4 month old male SHR and WKY, half of which received a single sc pellet of 12 mg estradiol benzoate and the remaining half a cholesterol implant. Hippocampi were dissected and processed for aromatase mRNA expression using real time PCR. A second batch of animals was processed for aromatase and glial fibrillary acidic protein (GFAP) immunocytochemistry. Basal level of aromatase mRNA was higher in SHR respect of WKY. Following estradiol treatment, aromatase mRNA was further increased in the SHR group only. In the hilus of the dentate gyrus of cholesterol-implanted SHR, we found aromatase immunoreactive cell processes and fibers more strongly stained respect of WKY rats. Estradiol treatment of SHR further increased the length of immunoreactive processes and fibers in the hilar region and also increased aromatase immunoreactivity in the CA1 but not the CA3 pyramidal cell region. WKY rats were spared from the estradiol effect. Double-labelling experiments showed that aromatase+ processes and fibers of the hilus of SHR-treated rats did no colocalize with GFAP+ astrocyte cell bodies or processes. In conclusion, basal and estradiol-stimulated aromatase expression was enhanced in hypertensive rat hippocampus. A combination of exogenous estrogens and those locally synthesized may better alleviate hypertensive encephalopathy.


Aromatase/biosynthesis , Estradiol/pharmacology , Estrogens/pharmacology , Hippocampus/drug effects , Animals , Aromatase/genetics , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/anatomy & histology , Hippocampus/metabolism , Immunohistochemistry , Male , Nerve Fibers/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Species Specificity
8.
J Neuroendocrinol ; 22(10): 1082-92, 2010 Oct.
Article En | MEDLINE | ID: mdl-20722975

The hippocampus of spontaneously hypertensive rats (SHR) and deoxycorticosterone (DOCA)-salt hypertensive rats shows decreased cell proliferation and astrogliosis as well as a reduced number of hilar cells. These defects are corrected after administration of 17ß-oestradiol (E(2) ) for 2 weeks. The present work investigated whether E(2) treatment of SHR and of hypertensive DOCA-salt male rats modulated the expression of brain-derived neurotrophic factor (BDNF), a neurotrophin involved in hippocampal neurogenesis. The neurogenic response to E(2) was simultaneously determined by counting the number of doublecortin-immunopositive immature neurones in the subgranular zone of the dentate gyrus. Both hypertensive models showed decreased expression of BDNF mRNA in the granular zone of the dentate gyrus, without changes in CA1 or CA3 pyramidal cell layers, decreased BDNF protein levels in whole hippocampal tissue, low density of doublecortin (DCX)-positive immature neurones in the subgranule zone and decreased length of DCX+ neurites in the dentate gyrus. After s.c. implantation of a single E(2) pellet for 2 weeks, BDNF mRNA in the dentate gyrus, BDNF protein in whole hippocampus, DCX immunopositive cells and the length of DCX+ neurites were significantly raised in both SHR and DOCA-salt-treated rats. These results indicate that: (i) low BDNF expression and deficient neurogenesis distinguished the hippocampus of SHR and DOCA-salt hypertensive rats and (ii) E(2) was able to normalise these biologically important functions in the hippocampus of hypertensive animals.


Brain-Derived Neurotrophic Factor/metabolism , Estradiol/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hypertension/physiopathology , Neurogenesis/physiology , Neuroprotective Agents/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Desoxycorticosterone/metabolism , Doublecortin Domain Proteins , Doublecortin Protein , Hippocampus/cytology , Hippocampus/pathology , Humans , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mineralocorticoids/metabolism , Neurons/cytology , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley
9.
Horm Metab Res ; 40(4): 245-50, 2008 Apr.
Article En | MEDLINE | ID: mdl-18548383

Using both IN VITRO and IN VIVO approaches, we studied the antagonism exerted by the synthetic progestin levonorgestrel on estrogen-induced prolactinomas, considering that levonorgestrel shows partial androgenic properties and that androgens inhibit estrogen-induced prolactin synthesis and secretion. In the tumors, binding of estrogens to their receptors was competed neither by progesterone receptor ligands nor by androgen receptor ligands, ruling out direct inhibitory effects of these drugs on tumor development. Progestin binding was competed by the progesterone receptor agonists progesterone and levonorgestrel, by the antagonist mifepristone, and also by the androgen dihydrotestosterone, whereas the androgen receptor antagonist hydroxyflutamide was a weak competitor. In addition, both progesterone receptor and androgen receptor ligands competed for binding to androgen receptors. In primary cultures of pituitary tumors, levonorgestrel decreased prolactin secretion, an effect that was blocked by mifepristone but not by hydroxyflutamide. IN VIVO results indicated that levonorgestrel inhibition of both estrogen-induced pituitary weight increment and hyperprolactinemia was reduced by mifepristone, whereas flutamide was unable to block levonorgestrel effects. Our results suggest that even when an interaction of levonorgestrel with androgen receptors in the tumors is possible, the antagonistic effects of levonorgestrel on tumor development and functionality are mediated by progesterone receptors.


Carcinogens , Diethylstilbestrol , Levonorgestrel/pharmacology , Pituitary Neoplasms/chemically induced , Pituitary Neoplasms/prevention & control , Progesterone Congeners/pharmacology , Receptors, Progesterone/antagonists & inhibitors , Animals , Binding, Competitive/drug effects , Hormone Antagonists/pharmacology , Male , Mifepristone/pharmacology , Organ Size/drug effects , Ovariectomy , Pituitary Gland, Anterior/drug effects , Prolactin/metabolism , Rats , Rats, Inbred F344 , Receptors, Androgen/drug effects
10.
J Neuroendocrinol ; 18(6): 466-74, 2006 Jun.
Article En | MEDLINE | ID: mdl-16684136

Hippocampal neuropathology is a recognised feature of the brain in spontaneously hypertensive rats (SHR), but similar studies are lacking in another model of hypertension, the mineralocorticoid-salt-treated rat. The present study aimed to compare changes in hippocampal parameters in 16-week-old male SHR (blood pressure approximately 190 mmHg) and their normotensive Wistar-Kyoto controls, with those of male Sprague-Dawley rats receiving (i) 10 mg deoxycorticosterone acetate (DOCA) every other day during 3 weeks and drinking 1% NaCl solution (blood pressure approximately 160 mmHg) and normotensive controls treated with (ii) DOCA and drinking water, (iii) drinking water only or (iv) 1% NaCl only. In these experimental groups, we determined: (i) cell proliferation in the dentate gyrus (DG) using the 5-bromo-2'-deoxyuridine-labelling technique; (ii) the number of glial fibrillary acidic protein (GFAP) positive astrocytes under the CA1, CA3 and DG; (iii) the number of apolipoprotein E (ApoE) positive astrocytes as a marker of potential neuronal damage; and (iv) the number of neurones in the hilus of the DG, taken as representative of neuronal density in other hippocampal subfields. Changes were remarkably similar in both models, indicating a decreased cell proliferation in DG, an increased number of astrocytes immunopositive for GFAP and ApoE and a reduced number of hilar neurones. Although hypertension may be a leading factor for these abnormalities, endocrine mechanisms may be involved, because hypothalamic-pituitary function, mineralocorticoid receptors and sensitivity to mineralocorticoid treatment are stimulated in SHR, whereas high exogenous mineralocorticoid levels circulate in DOCA-treated rats. Thus, in addition to the deleterious effects of hypertension, endocrine factors may contribute to the abnormalities of hippocampus in SHR and DOCA-treated rats.


Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Hypertension/pathology , Hypertension/physiopathology , Animals , Apolipoproteins E/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Cell Division , Desoxycorticosterone/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Gliosis/physiopathology , Male , Neurons/pathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Sodium Chloride/pharmacology , Species Specificity
11.
J Neuroendocrinol ; 16(1): 64-71, 2004 Jan.
Article En | MEDLINE | ID: mdl-14962078

We have previously described a sexual dimorphism in oestrogen-induced anterior pituitary tumorigenesis in Fischer 344 rats, with female tumours averaging twice the size of those of males. Neonatal androgenization of female Fischer 344 rats with 100 micro g of testosterone propionate reverted that effect, causing a 'male-like' phenotype. The peptides galanin and vasoactive intestinal peptide (VIP) are possible mediators of oestrogen effects on the anterior pituitary, including hyperprolactinemia and lactotroph proliferation. To further extend our previous findings, we investigated the expression of galanin and VIP in the anterior pituitary of control and oestrogenized male, female and neonatally androgenized female Fischer 344 rats. At 3 months of age, rats were deprived of their gonads and divided into control and diethylstilbestrol (DES)-treated groups. In the anterior pituitary of control rats, galanin and VIP immunoreactive cells were absent. However, in DES-treated rats, pituitaries from normal ovariectomized females showed higher number of galanin and VIP positive cells than pituitaries from neonatally androgenized ovariectomized females and gonadectomized males. This pattern correlated with changes in anterior pituitary weight and serum prolactin. Our study suggests that sexual differences in oestrogen-induced pituitary tumorigenesis could be due to the differential expression of galanin and VIP. Furthermore, our data support the fact that neonatal exposure to androgens, as in normal males and androgenized females, may condition the response of the pituitary gland to oestrogens in adult life.


Galanin/metabolism , Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Sex Differentiation/physiology , Vasoactive Intestinal Peptide/metabolism , Animals , Animals, Newborn , Diethylstilbestrol , Female , Galanin/genetics , Male , Pituitary Neoplasms/chemically induced , Pituitary Neoplasms/pathology , Prolactinoma/chemically induced , Prolactinoma/pathology , Rats , Rats, Inbred F344 , Sex Characteristics , Testosterone , Virilism/chemically induced
12.
Neuroendocrinology ; 74(6): 396-406, 2001 Dec.
Article En | MEDLINE | ID: mdl-11752896

Salt appetite, a conditioning factor for hypertension and cardiovascular diseases, is produced when high doses of mineralocorticoids are given to experimental animals. A commonly used procedure to identify neuronal activation is to determine the number of Fos-immunoreactive cells. In rats with established salt appetite after 8 days of deoxycorticosterone acetate (DOCA) treatment, Fos-positive cells were studied in seven brain areas. Significant increases in Fos activity were recorded in the paraventricular (PVN) and supraoptic (SON) nuclei, median preoptic nucleus (MnPO), organum vasculosum of the lamina terminalis (OVLT), preoptic area (POA), bed nucleus of the stria terminalis (BNST) and amygdala (AMYG). In most of these areas, increased Fos expression was also observed early (2 h) after a single DOCA injection, well before salt appetite develops. Using a mineralocorticoid receptor (MR) antibody, we studied whether Fos-active regions also expressed MR. MR-positive cells were found in the OVLT, MnPO, AMYG and BNST, but not in the POA, PVN and SON. In the PVN and SON, nevertheless, prolonged or single DOCA treatment increased expression of mRNA for arginine vasopressin (AVP). The present demonstration of Fos activation, in conjunction with differential expression of MR and stimulation of AVP mRNA, suggests that a neuroanatomical pathway comprising the AMYG, osmosensitive brain regions and magnocellular nuclei becomes activated during DOCA effects on salt appetite. It is recognized, however, that DOCA effects may also depend on mechanisms and brain structures other than those considered in the present investigation. Since some Fos-positive regions were devoid of MR, a comprehensive view of DOCA-induced salt appetite should consider nongenomic pathways of steroid action, including the role of reduced DOC metabolites binding to GABAergic membrane receptors.


Appetite/physiology , Brain/metabolism , Desoxycorticosterone/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Sodium Chloride , Animals , Arginine Vasopressin/genetics , Immunohistochemistry , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/metabolism , Tissue Distribution
13.
J Neuroendocrinol ; 13(3): 302-9, 2001 Mar.
Article En | MEDLINE | ID: mdl-11207946

Galanin is a peptide widely distributed in the hypothalamic-pituitary axis. In the female rat pituitary, galanin is mainly present in lactotrophs, where it regulates their secretion and proliferation. Galanin expression is increased in oestrogen-induced prolactinomas, and it has been proposed that oestrogen effects on lactotroph function and proliferation could be mediated by galanin. Previous studies from our laboratory demonstrated that the synthetic progestin levonorgestrel antagonizes pituitary tumorigenesis of rats given oestrogen, reducing the number of proliferating cells and increasing cell death by nonapoptotic mechanism(s). To elucidate the role of galanin in levonorgestrel effects on the tumours, we examined galanin and prolactin mRNA and peptide expression in prolactinomas of rats receiving the progestin. Levonorgestrel reduced the pituitary weight and serum prolactin concentrations in oestrogen-treated rats. Galanin mRNA expression (determined by in situ hybridization), and the number of galanin expressing cells (determined by immunocytochemistry) were also reduced by the progestin in tumour-bearing rats. However, neither prolactin mRNA content, nor the number of prolactin-expressing cells, were modified by levonorgestrel treatment of oestrogen-receiving rats. The present study suggests that levonorgestrel controls pituitary growth by diminishing galanin expression. In contrast, changes in serum prolactin concentration seem to be more related to the reduction in tumour size, since the reduction in galanin expression was not large enough to regulate prolactin mRNA expression or the percentage of lactotrophs.


Diethylstilbestrol , Galanin/genetics , Levonorgestrel/pharmacology , Pituitary Neoplasms/metabolism , Progesterone Congeners/pharmacology , Prolactin/genetics , Animals , Cell Count , Female , Galanin/analysis , Gene Expression/drug effects , Immunohistochemistry , In Situ Hybridization , Organ Size/drug effects , Ovariectomy , Pituitary Gland/pathology , Pituitary Neoplasms/chemically induced , Pituitary Neoplasms/pathology , Prolactin/analysis , Prolactin/blood , RNA, Messenger/analysis , Rats , Rats, Inbred F344
14.
Neuroendocrinology ; 72(2): 80-90, 2000 Aug.
Article En | MEDLINE | ID: mdl-10971143

Female F344 rats treated chronically with diethylstilbestrol (DES) develop prolactin (PRL)-producing pituitary tumors. These tumors are larger in female than in male rats. To investigate gender differences in DES-induced pituitary tumor formation, we employed female and male rats and neonatally androgenized females, which received 100 microg of testosterone propionate (TP) after birth. At 3 months of age, all rats were deprived of their gonads and divided into control and DES-treated groups. Forty days after beginning treatment, control pituitary weight and serum PRL were similar in gonadectomized males (GDX), ovariectomized females (OVX) and androgenized-ovariectomized females (OVX + TP), but weight of DES-induced tumors was 2.5-fold higher and serum PRL 5.6-fold higher in OVX + DES than in GDX + DES or OXV + TP + DES (p<0.001). At the pituitary level, nuclear estrogen receptors (NE(2)R) amounted to >100 fmol/mg DNA in all rats receiving DES. However, NE(2)R were lower in OVX + DES (101.3+/-9.0 fmol/mg DNA) than in GDX + DES (174.6 +/-16.8; p<0.05) and in OXV + DES + TP (150.3+/-27.7; p<0.05). A similar profile was found for cytosolic progestin receptors. Using electron microscopy (EM), hyperplasia/hypertrophy of lactotropes was found in all DES-stimulated pituitaries. However, tumors of OVX + DES rats were enriched in hyperstimulated typical lactotropes, i.e., cells with high rate of hormonal synthesis, processing and secretion. Instead, tumors from GDX + DES and OVX + TP + DES rats were a mixture of typical and atypical lactotropes, i.e. a cell subpopulation with refractory secretory response and a few gonadotropes. In agreement with these data, immunoreactive pituitary PRL was lower in OVX + DES than in OVX + TP + DES and GDX + DES groups. Thus, differences in the sensitivity to DES, serum and tumor PRL, NE(2)R and progestin receptors between estrogenized female rats on one side and male and TP-androgenized females on the other, may by due in part to heterogeneity of cell populations. Our data further suggest that neonatal hypothalamic exposure to androgens, as in normal males or androgenized females with masculinization of hypothalamic centers, may condition the response to DES stimulation later in life.


Carcinogens/toxicity , Diethylstilbestrol/toxicity , Pituitary Neoplasms/chemically induced , Pituitary Neoplasms/pathology , Prolactin/metabolism , Prolactinoma/chemically induced , Prolactinoma/pathology , Animals , Cytosol/metabolism , Diethylstilbestrol/blood , Female , Male , Microscopy, Electron , Orchiectomy , Organ Size/physiology , Ovariectomy , Pituitary Neoplasms/metabolism , Prolactin/blood , Prolactinoma/metabolism , Rats , Rats, Inbred F344 , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Receptors, Steroid/metabolism , Sex Characteristics
...