Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
1.
Am J Physiol Heart Circ Physiol ; 318(1): H34-H48, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31675258

ABSTRACT

Obstructive sleep apnea is characterized by interrupted breathing that leads to cardiovascular sequelae including chronic hypertension that can persist into the waking hours. Chronic intermittent hypoxia (CIH), which models the hypoxemia associated with sleep apnea, is sufficient to cause a sustained increase in blood pressure that involves the central nervous system. The median preoptic nucleus (MnPO) is an integrative forebrain region that contributes to blood pressure regulation and neurogenic hypertension. The MnPO projects to the paraventricular nucleus (PVN), a preautonomic region. We hypothesized that pathway-specific lesions of the projection from the MnPO to the PVN would attenuate the sustained component of chronic intermittent hypoxia-induced hypertension. Adult male Sprague-Dawley rats (250-300 g) were anesthetized with isoflurane and stereotaxically injected bilaterally in the PVN with a retrograde Cre-containing adeno-associated virus (AAV; AAV9.CMV.HI.eGFP-Cre.WPRE.SV40) and injected in the MnPO with caspase-3 (AAV5-flex-taCasp3-TEVp) or control virus (AAV5-hSyn-DIO-mCherry). Three weeks after the injections the rats were exposed to a 7-day intermittent hypoxia protocol. During chronic intermittent hypoxia, controls developed a diurnal hypertension that was blunted in rats with caspase lesions. Brain tissue processed for FosB immunohistochemistry showed decreased staining with caspase-induced lesions of MnPO and downstream autonomic-regulating nuclei. Chronic intermittent hypoxia significantly increased plasma levels of advanced oxidative protein products in controls, but this increase was blocked in caspase-lesioned rats. The results indicate that PVN-projecting MnPO neurons play a significant role in blood pressure regulation in the development of persistent chronic intermittent hypoxia hypertension.NEW & NOTEWORTHY Chronic intermittent hypoxia associated with obstructive sleep apnea increases oxidative stress and leads to chronic hypertension. Sustained hypertension may be mediated by angiotensin II-induced neural plasticity of excitatory median preoptic neurons in the forebrain that project to the paraventricular nucleus of the hypothalamus. Selective caspase lesions of these neurons interrupt the drive for sustained hypertension and cause a reduction in circulating oxidative protein products. This indicates that a functional connection between the forebrain and hypothalamus is necessary to drive diurnal hypertension associated with intermittent hypoxia. These results provide new information about central mechanisms that may contribute to neurogenic hypertension.


Subject(s)
Apoptosis , Arterial Pressure , Caspase 3/metabolism , Hypertension/prevention & control , Hypoxia/complications , Paraventricular Hypothalamic Nucleus/enzymology , Preoptic Area/enzymology , Animals , Caspase 3/genetics , Circadian Rhythm , Disease Models, Animal , Heart Rate , Hypertension/enzymology , Hypertension/pathology , Hypertension/physiopathology , Hypoxia/enzymology , Hypoxia/pathology , Hypoxia/physiopathology , Male , Oxidative Stress , Paraventricular Hypothalamic Nucleus/pathology , Paraventricular Hypothalamic Nucleus/physiopathology , Preoptic Area/pathology , Preoptic Area/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Signal Transduction
2.
Cereb Cortex ; 27(12): 5696-5714, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29117290

ABSTRACT

The proliferative niches in the subpallium generate a rich cellular variety fated for diverse telencephalic regions. The embryonic preoptic area (POA) represents one of these domains giving rise to the pool of cortical GABAergic interneurons and glial cells, in addition to striatal and residual POA cells. The migration from sites of origin within the subpallium to the distant targets like the cerebral cortex, accomplished by the adoption and maintenance of a particular migratory morphology, is a critical step during interneuron development. To identify factors orchestrating this process, we performed single-cell transcriptome analysis and detected Dnmt1 expression in murine migratory GABAergic POA-derived cells. Deletion of Dnmt1 in postmitotic immature cells of the POA caused defective migration and severely diminished adult cortical interneuron numbers. We found that DNA methyltransferase 1 (DNMT1) preserves the migratory shape in part through negative regulation of Pak6, which stimulates neuritogenesis at postmigratory stages. Our data underline the importance of DNMT1 for the migration of POA-derived cells including cortical interneurons.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/embryology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Interneurons/enzymology , Neural Stem Cells/enzymology , Preoptic Area/embryology , Animals , Animals, Newborn , Cell Count , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , DNA Methylation , GABAergic Neurons/cytology , GABAergic Neurons/enzymology , Interneurons/cytology , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/cytology , Neuronal Outgrowth/physiology , Preoptic Area/cytology , Preoptic Area/enzymology , Tissue Culture Techniques , Transcriptome , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
3.
Horm Behav ; 93: 137-150, 2017 07.
Article in English | MEDLINE | ID: mdl-28576650

ABSTRACT

Since Arnold Adolph Berthold established in 1849 the critical role of the testes in the activation of male sexual behavior, intensive research has identified many sophisticated neurochemical and molecular mechanisms mediating this action. Studies in Japanese quail demonstrated the critical role of testosterone action and of testosterone aromatization in the sexually dimorphic medial preoptic nucleus in the activation of male copulatory behavior. The development of an immunohistochemical visualization of brain aromatase in quail then allowed further refinement in the localization of the sites of neuroestrogens production. Testosterone aromatization is required for the activation of both appetitive and consummatory aspects of male sexual behavior. Brain aromatase activity is modulated by steroid-induced changes in the transcription of the corresponding gene but also more rapidly by phosphorylation processes. Sexual interactions with a female also rapidly regulate brain aromatase activity in an anatomically specific manner presumably via the release and action of endogenous glutamate. These rapid changes in estrogen production modulate sexual behavior and in particular its motivational component with latencies ranging between 15 and 30min. Brain estrogens seem to act in a manner akin to a neurotransmitter or at least a neuromodulator. More recently, assays of brain estradiol concentrations in micropunched samples or in dialysis samples obtained from behaviorally active males suggested that aromatase activity measured ex vivo might not be an accurate proxy to the rapid changes in local neuroestrogens production and concentrations. Studies of brain testosterone metabolism are thus not over and will keep scientists busy for a little longer. Elsevier SBN Keynote Address, Montreal.


Subject(s)
Aromatase/metabolism , Brain/enzymology , Estrogens/metabolism , Masculinity , Sexual Behavior , Testosterone/metabolism , Animals , Coturnix , Estrogen Receptor beta/metabolism , Female , Humans , Male , Models, Animal , Preoptic Area/enzymology , Social Behavior
4.
J Exp Biol ; 219(Pt 24): 3861-3865, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27742893

ABSTRACT

Dopamine regulates reproduction in part by modulating neuronal activity within the hypothalamic-pituitary-gonadal (HPG) axis. Previous studies suggested numerous mechanisms by which dopamine exerts inhibitory control over the HPG axis, ultimately changing the levels of sex steroids that regulate reproductive behaviors. However, it is not known whether these mechanisms are conserved across vertebrate species. In particular, it is unknown whether mechanisms underlying dopaminergic control of reproduction are shared between mammals and teleost fish. In mammals, dopamine directly inhibits gonadotropin-releasing hormone (GnRH1) hypothalamic neurons, the gatekeepers for activation of the HPG axis. Here, we demonstrate, for the first time in teleost fish, dopaminergic control of GnRH1 neurons via direct dopamine type-2-like receptor (D2R)-mediated inhibition within the hypothalamus. These results suggest that direct dopaminergic control of GnRH1 neurons via interactions in the hypothalamus is not exclusive to tetrapod reproductive control, but is likely conserved across vertebrate species.


Subject(s)
Cichlids/physiology , Dopamine/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Female , Male , Neurons/drug effects , Preoptic Area/drug effects , Preoptic Area/enzymology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Sex Characteristics , Tyrosine 3-Monooxygenase/metabolism
5.
Clin Exp Pharmacol Physiol ; 43(10): 960-6, 2016 10.
Article in English | MEDLINE | ID: mdl-27297082

ABSTRACT

Previous reports indicate that overexpression of copper/zinc superoxide dismutase (CuZnSOD), an intracellular superoxide (O2 (•-) ) scavenging enzyme, in the brain subfornical organ improves cardiac function in a mouse model of heart failure (HF). A downstream hypothalamic site, the MnPO, may act as a relay centre for O2 (•-) to serve as a mediator in the pathophysiology of HF. To test the hypothesis that elevated O2 (•-) in the MnPO contributes to the pathophysiology of HF and decreased cardiac function, we injected adenovirus encoding CuZnSOD (AdCuZnSOD, n=7) or control empty adenovirus vector (AdEmpty, n=7) into the MnPO of normal rats. Subsequently, rats were subjected to coronary artery ligation to create a myocardial infarct (MI) of the left ventricle. Cardiac function was monitored via echocardiography. Upon completion, rat brains were examined for CuZnSOD expression in MnPO via immunofluorescence and histopathological analyses of cardiac infarct size were conducted. Baseline (EF) ejection fractions (%) of AdCuZnSOD and AdEmpty rats were 73 ± 1 and 71 ± 1, respectively. Two weeks after MI, EF was significantly decreased in both groups of rats (AdCuZnSOD: 51 ± 3, AdEmpty: 46 ± 1). In contrast, by 4 weeks post MI, EF had improved to 64 ± 2 in AdCuZnSOD rats, yet was only 52 ± 1 in AdEmpty rats, and this was accompanied by lower plasma noradrenaline levels in AdCuZnSOD rats (0.49 ± 0.19 ng/mL) compared to AdEmpty rats (1.20 ± 0.32 ng/mL). In conclusion, despite decreases in EF early after MI, overexpression of CuZnSOD in the MnPO was related to an improvement in left ventricular function and concomitant decreased plasma noradrenaline levels 4 weeks post MI.


Subject(s)
Gene Expression Regulation, Enzymologic , Myocardial Infarction/enzymology , Preoptic Area/enzymology , Superoxide Dismutase/biosynthesis , Ventricular Function, Left/physiology , Animals , Male , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/genetics
6.
Gen Comp Endocrinol ; 208: 64-72, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25157789

ABSTRACT

Various studies in rodents recently concluded that puberty should be considered as a second period of organization of brain and behavior and that action of sex steroids at that time is long lasting and possibly permanent. We tested this notion in male Japanese quail that had been castrated before 3weeks post-hatch by analyzing whether a similar treatment with exogenous testosterone initiated at 3, 5 or 7weeks post-hatch has a differential influence on the development of testosterone-dependent morphological, behavioral and neural characteristics that are known to be sexually differentiated. The growth of the androgen-dependent cloacal gland was significantly faster when testosterone treatment was initiated later in life indicating that the target tissue is not ready to fully respond to androgens at 3weeks post-hatch. The three groups of birds nevertheless developed a gland of the same size typical of intact sexually mature birds. When adults, all birds expressed copulatory behavior with the same frequencies and latencies and they displayed the same level of aromatase activity and of vasotocinergic innervation in the preoptic area as gonadally intact males despite the fact that they had been treated with testosterone for different durations starting at different ages. Surprisingly, the frequency of cloacal sphincter contractions, a measure of appetitive sexual behavior, was significantly higher when testosterone treatment had been initiated later. Together these data provide no clear evidence for an organizational action of testosterone during sexual maturation of male quail but additional experiments should investigate whether estrogens have such an action in females.


Subject(s)
Aging/physiology , Coturnix/physiology , Testosterone/pharmacology , Aging/drug effects , Animals , Aromatase/metabolism , Body Weight/drug effects , Female , Male , Preoptic Area/drug effects , Preoptic Area/enzymology , Sexual Behavior, Animal/drug effects , Sexual Maturation/drug effects , Vasotocin/metabolism
7.
Dev Neurosci ; 34(4): 318-26, 2012.
Article in English | MEDLINE | ID: mdl-22832237

ABSTRACT

Fever is a major component of the host's defense against infection. Inadequate febrile response can predispose an individual to the deleterious effects of infection. Neonatal exposure to infectious agents such as bacterial lipopolysaccharide (LPS) permanently dampens the adult febrile response. Whether prenatal immune challenge alters febrile response during adulthood is still not known. In the present study, LPS (100 µg/kg, i.p.) or pyrogen-free saline was administered to pregnant rats on either gestation day (GD) 12, 15 or 19 and the febrile response of their respective adult offspring was monitored. During adulthood (>70 days old), the rats born to LPS-injected dams on GD15 displayed a significantly attenuated febrile response to LPS (50 µg/kg, i.p.) compared to their control counterparts born to dams given saline on GD15. Immune challenge during either early (GD12) or late (GD19) pregnancy did not have a significant impact on fever in the adult offspring. Immune challenge on GD15, but not on GD12 or 19, heightened the plasma corticosterone response to a subsequent LPS injection to the adult offspring but did not have a significant effect on their basal plasma corticosterone levels. Finally, LPS-induced COX-2 in the fever-controlling regions of the hypothalamus was significantly reduced in the adult rats born to dams given LPS on GD15 compared to their counterparts born to dams given saline on GD15. Such COX-2 reduction was not observed in the adult offspring born to dams given LPS on either GD12 or 19. Taken together, these data suggest that a single immune challenge during a critical window of pregnancy alters the neuroimmune response in adult offspring.


Subject(s)
Endotoxemia/physiopathology , Fever/physiopathology , Hypothalamo-Hypophyseal System/embryology , Pituitary-Adrenal System/embryology , Pregnancy Complications, Infectious/physiopathology , Prenatal Exposure Delayed Effects , Animals , Body Temperature/drug effects , Body Temperature/physiology , Corticosterone/blood , Cyclooxygenase 2/analysis , Dose-Response Relationship, Drug , Endotoxemia/immunology , Enzyme Induction/drug effects , Female , Fever/blood , Fever/chemically induced , Gestational Age , Hypothalamo-Hypophyseal System/physiopathology , Lipopolysaccharides/toxicity , Male , Pituitary-Adrenal System/physiopathology , Pregnancy , Preoptic Area/embryology , Preoptic Area/enzymology , Rats , Rats, Sprague-Dawley
8.
Gen Comp Endocrinol ; 170(3): 475-9, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21074531

ABSTRACT

CYP17A1 encodes the key enzyme of androgen biosynthesis, P450c17. The gene is expressed in a number of steroidogenic tissues among them testis, ovary, placenta and adrenal gland. The proper analysis of CYP17A1 expression and of epigenetic parameters however, is hampered by the presence of more than one copy of the gene within the bovine genome. Therefore, as a prerequisite for future studies we characterized these copies and analyzed their promoter methylation and expression profiles in different tissues. DNA methylation levels were determined by bisulfite modification, amplification, cloning and sequencing. Transcription was analyzed by RT-PCR. From bovine genomic DNA three different CYP17A1 promoter sequences could be amplified with a sequence similarity of 94.8%, 95.6% and 98.7%. Based on these sequences we could reconstruct, by in silico analysis, the promoter regions and eight potentially coding exons of two loci, CYP17A1a and CYP17A1b, and the promoter region and truncated first exon of a third locus, CYP17A1x. By using locus-specific primers, only transcripts of CYP17A1a, but not of CYP17A1b could be detected in testis, epididymis, theca, corpus luteum, placental cotyledons, adrenal gland and preoptic brain area. Methylation analysis revealed that only the CYP17A1a promoter was hypo-methylated in the tested P450c17 active tissues, whereas both other copies showed higher levels of methylation. From these data we conclude that the bovine genome contains three paralogous copies of the CYP17A1 gene, of which two (CYP17A1b and CYP17A1x) might be silenced by epigenetic modification (promoter methylation).


Subject(s)
Cattle/genetics , Steroid 17-alpha-Hydroxylase/genetics , Animals , Corpus Luteum/enzymology , DNA Methylation , Epididymis/enzymology , Epigenomics , Female , Male , Placenta/enzymology , Pregnancy , Preoptic Area/enzymology , Promoter Regions, Genetic , Testis/enzymology , Theca Cells/enzymology
9.
J Neurosci ; 29(31): 9714-8, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19657024

ABSTRACT

The sexually dimorphic population of dopamine neurons in the anteroventral periventricular nucleus of the preoptic region of the hypothalamus (AVPV) develops postnatally under the influence of testosterone, which is aromatized to estrogen. There are fewer dopaminergic neurons labeled with tyrosine hydroxylase (TH) in the male AVPV than the female, and sex steroids determine this sex difference, yet the role of cell death in specifying numbers of dopaminergic neurons in the AVPV is unknown. Estradiol treatment of the AVPV, in vivo and in vitro, was used to manipulate TH-ir cell number. In vitro, concurrent treatment with the estrogen receptor antagonist ICI 182,780 rescued TH-ir cells. Cyclosporin A, an inhibitor of cell death dependent on the opening of a mitochondrial permeability transition pore also blocked TH-ir cell loss. In vivo, estradiol increased the number of apoptotic profiles, both TUNEL and Hoechst labeled nuclei, in the AVPV. This increased apoptosis was also dependent on the presence of the alpha form of the estrogen receptor. To test for caspase dependent TH-ir cell loss, the pancaspase inhibitor ZVAD (N-benzyloxycabonyl-Val-Ala-Asp-fluoromethylketone) was used to rescue TH-ir cells from estradiol-mediated reduction in number. Together, these data suggest that an intrinsic cell death pathway is activated by estrogen to regulate TH-ir cell number. Thus, in contrast to the more widespread neuroprotective actions of sex steroids in the mammalian nervous system, in the AVPV estrogen regulates dopaminergic neuron number through a caspase-dependent mechanism of apoptotic cell death.


Subject(s)
Caspases/metabolism , Estrogens/metabolism , Neurons/physiology , Preoptic Area/growth & development , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase Inhibitors , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Female , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Preoptic Area/drug effects , Preoptic Area/enzymology , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
10.
BMC Genet ; 11: 62, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20609214

ABSTRACT

BACKGROUND: Mutarotases are recently characterized family of enzymes that are involved in the anomeric conversions of monosaccharides. The mammalian fucose mutarotase (FucM) was reported in cultured cells to facilitate fucose utilization and incorporation into protein by glycosylation. However, the role of this enzyme in animal has not been elucidated. RESULTS: We generated a mutant mouse specifically lacking the fucose mutarotase (FucM) gene. The FucM knockout mice displayed an abnormal sexual receptivity with a drastic reduction in lordosis score, although the animals were fertile due to a rare and forced intromission by a typical male. We examined the anteroventral periventricular nucleus (AVPv) of the preoptic region in brain and found that the mutant females showed a reduction in tyrosine hydoxylase positive neurons compared to that of a normal female. Furthermore, the mutant females exhibited a masculine behavior, such as mounting to a normal female partner as well as showing a preference to female urine. We found a reduction of fucosylated serum alpha-fetoprotein (AFP) in a mutant embryo relative to that of a wild-type embryo. CONCLUSIONS: The observation that FucM-/- female mouse exhibits a phenotypic similarity to a wild-type male in terms of its sexual behavior appears to be due to the neurodevelopmental changes in preoptic area of mutant brain resembling a wild-type male. Since the previous studies indicate that AFP plays a role in titrating estradiol that are required to consolidate sexual preference of female mice, we speculate that the reduced level of AFP in FucM-/- mouse, presumably resulting from the reduced fucosylation, is responsible for the male-like sexual behavior observed in the FucM knock-out mouse.


Subject(s)
Carbohydrate Epimerases/physiology , Sexual Behavior, Animal , Animals , Female , Mice , Preoptic Area/enzymology , Tyrosine 3-Monooxygenase/metabolism
11.
Brain Behav Evol ; 76(3-4): 279-88, 2010.
Article in English | MEDLINE | ID: mdl-21116109

ABSTRACT

The 5α-reductase (5αR) enzyme converts testosterone to 5α-dihydrotestosterone. This local metabolism within the brain is important for the full expression of male sexual behavior in many species, including green anole lizards. Two isozymes of 5αR exist and little is known about their specific distributions. We conducted in situ hybridization for both isozymes in intact male and female green anole brains during the breeding (BS) and non-breeding (NBS) seasons. 5αR1 mRNA was only detected in the brainstem, while 5αR2 was expressed in specific areas throughout the brain. As our primary interest was evaluating the potential role of 5αR in forebrain regulation of reproductive behavior, we quantified 5αR2 expression in the preoptic area, amygdala (AMY), and ventromedial hypothalamus (VMH). More 5αR2 cells were detected during the NBS than BS in the AMY, and the density of these cells was greater in females than males. In the VMH, the right side contained more 5αR2 cells than the left, an effect driven by a lateralized increase in the NBS. These data expand understanding of the distribution and potential roles of both isozymes in the adult brain, and differences in expression patterns between mammals and birds suggest that they may have been co-opted for different functions later in evolution.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Lizards/metabolism , Prosencephalon/enzymology , Sexual Behavior, Animal/physiology , Amygdala/enzymology , Animals , Female , Functional Laterality , In Situ Hybridization , Isoenzymes , Male , Preoptic Area/enzymology , Seasons , Sex Factors , Tissue Distribution , Ventromedial Hypothalamic Nucleus/enzymology
12.
Physiol Behav ; 97(1): 30-5, 2009 Apr 20.
Article in English | MEDLINE | ID: mdl-19419676

ABSTRACT

In young black-headed gulls (Larus ridibundus), exposure to testosterone increases the sensitivity of agonistic behaviour to a subsequent exposure to this hormone. The aim of this paper is twofold: to analyze whether social experience, gained during testosterone exposure, mediates this increase in hormonal sensitivity (priming), and whether this in turn is mediated by an increase in central aromatase activity. To this end, we performed three experiments. In the first juvenile gulls were exposed to two consecutive treatments with testosterone (T1 and T2), with more than a week interval in between. During T1, half of the birds were housed in social isolation (Iso) and the other half in groups (Soc). All birds were re-housed in a new social situation during the second treatment. The increase in social behaviour during T2 was significantly more rapid in Soc than Iso birds. In experiment 2 we show that 17beta-estradiol treatment facilitates the behaviour measured in experiment 1. In experiment 3 we used a set-up comparable with that of experiment 1, but birds were sacrificed early in the T2 period. Aromatase activity in the preoptic area and the hypothalamus was measured using the tritiated water releasing method. In some parts of the preoptic area and hypothalamus aromatase activity was higher in Soc birds relative to Iso birds. The results indicate that social experience can modulate the increase of social behaviour to testosterone via modulation of aromatase activity and independently of actual hormone levels.


Subject(s)
Agonistic Behavior/drug effects , Aromatase/metabolism , Social Isolation , Testosterone/administration & dosage , Testosterone/pharmacology , Aging , Animals , Charadriiformes , Drug Implants , Estradiol/administration & dosage , Estradiol/blood , Estradiol/pharmacology , Female , Hypothalamus/enzymology , Male , Preoptic Area/enzymology , Testosterone/blood
13.
Zoolog Sci ; 26(2): 163-70, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19341335

ABSTRACT

We examined the effects of hyperosmotic stress on the gene expression and activity of neuronal nitric oxide synthase (nNOS) in the preoptic/hypothalamic neurosecretory system of the euryhaline tilapia Oreochromis mossambicus (Mozambique tilapia) by means of semiquantitative RT-PCR and NADPHd histochemistry. Expression of nos1 was rapidly and transiently up-regulated in the preoptic region and hypothalamus in response to a salinity change (70% seawater, SW). Expression levels increased 4 h after the salinity change and then returned to basal levels within 8 h of the hyperosmotic challenge. NADPHd histochemistry revealed that positive magnocellular and gigantocellular preoptic neurons increased in number 4 h after the salinity change, while the number of parvocellular preoptic neurons reactive for NADPHd showed no significant change. These results indicate that the nNOS gene expression and NOS activity are stimulated in the preoptic/ hypothalamic neurosecretory system in response to hyperosmotic stress and suggest that NO influences neuronal responses to short-term osmotic stimulation in euryhaline fish.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Hypothalamo-Hypophyseal System/enzymology , Nitric Oxide Synthase Type I/biosynthesis , Preoptic Area/enzymology , Tilapia/physiology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Histocytochemistry , Male , Molecular Sequence Data , NADPH Dehydrogenase/chemistry , Nitric Oxide Synthase Type I/genetics , Osmotic Pressure , Phylogeny , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Tilapia/genetics , Tilapia/metabolism
14.
Psychopharmacology (Berl) ; 236(4): 1293-1301, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30539267

ABSTRACT

RATIONALE: Lycium barbarum polysaccharide (LBP) is known to promote reproductive functions. However, its role in noncontact erection (NCE) of penis initiated by brain regions including medial preoptic area (MPOA) and paraventricular nucleus (PVN) regions responsible for sexual behavior has not been investigated. OBJECTIVES: Therefore, this study initially investigated the effects of LBP on male sexual function, and subsequently, the mechanistic insight was investigated through assessing the expression of neuronal nitric oxide synthase (nNOS) in the MPOA and PVN. METHODS: The adult male rats were treated with 100 mg/kg of LBP or vehicle by oral gavage. Before and after 14 days of treatment, copulatory behavior and noncontact erection (NCE) were recorded. After the last behavioral test, the brain was isolated to measure nNOS expression in the MPOA and PVN. RESULTS: Data showed that LBP treatment significantly increased both the frequencies of intromission as well as ejaculation, compared to the control group. Whereas, a reduced post-ejaculatory interval was observed compared to same group on day 0. Furthermore, the treatment led to an increased intromission ratio, inter-intromission interval, and the number of MPOA nNOS-immunoreactive cells (nNOS-ir). Additionally, a significantly positive correlation between ejaculation frequency and MPOA nNOS-ir cells was recorded. Of note, LBP treatment had no effects on NCE and PVN nNOS-ir expression. CONCLUSION: These findings suggest that LBP enhances sexual behavior through increased nNOS expression in the MPOA in male rats.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Nitric Oxide Synthase Type I/metabolism , Penile Erection/drug effects , Preoptic Area/drug effects , Sexual Behavior, Animal/drug effects , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Male , Neurons/drug effects , Neurons/enzymology , Nitric Oxide , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/enzymology , Penile Erection/physiology , Preoptic Area/enzymology , Rats , Rats, Long-Evans , Sexual Behavior, Animal/physiology , Testis/drug effects , Testis/enzymology
15.
Endocrinology ; 149(11): 5518-26, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18617607

ABSTRACT

In addition to the activation of classical progestin receptor-dependent genomic pathway, progesterone (P) can activate nonclassical, membrane-initiated signaling pathways in the brain. We recently demonstrated rapid P activation of second-messenger kinases, protein kinase A, and protein kinase C in the ventromedial nucleus (VMN) and preoptic area (POA) of rat brain. To determine whether P can activate yet another Ca+2 dependent kinase, we examined the rapid P modulation of calcium and calmodulin-dependent protein kinase II (CaMKII) in the VMN and POA in female rats. A rapid P-initiated activation of CaMKII basal activity was observed in the VMN but not the POA at 30 min. Estradiol benzoate (EB) priming enhanced this CaMKII basal activity in both the VMN and POA. CaMKII protein levels and phosphorylation of Thr-286 moiety on CaMKII, however, remained unchanged with EB and/or P treatments, suggesting that the changes in the CaMKII kinase activity are due to rapid P modulation of the kinase activity and not its synthesis or autoactivation. Furthermore, intracerebroventricular (icv) administration of a CaMKII-specific inhibitor, KN-93, 30 min prior to the P infusion, in EB-primed, ovariectomized female rats inhibited CaMKII activation but not protein kinase A and protein kinase C activities. Interestingly, icv administration of KN-93 30 min prior to P infusion (icv) resulted in a reduction but not total inhibition of P-facilitated lordosis response in EB-primed female rats. These observations suggest a redundancy or, alternately, a hierarchy in the P-regulated activation of kinase signaling cascades in female reproductive behavior.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Hypothalamus/drug effects , Hypothalamus/enzymology , Progesterone/pharmacology , Animals , Benzylamines/pharmacology , Brain/drug effects , Brain/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Enzyme Activation/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Hypothalamus/metabolism , Ovariectomy , Phosphorylation/drug effects , Preoptic Area/drug effects , Preoptic Area/enzymology , Preoptic Area/metabolism , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Sexual Behavior, Animal/drug effects , Signal Transduction/drug effects , Sulfonamides/pharmacology
16.
Endocrinology ; 149(11): 5509-17, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18617608

ABSTRACT

The modulation of gene regulation by progesterone (P) and its classical intracellular regulation by progestin receptors in the brain, resulting in alterations in physiology and behavior has been well studied. The mechanisms mediating the short latency effects of P are less well understood. Recent studies have revealed rapid nonclassical signaling action of P involving the activation of intracellular signaling pathways. We explored the involvement of protein kinase C (PKC) in P-induced rapid signaling in the ventromedial nucleus of the hypothalamus (VMN) and preoptic area (POA) of the rat brain. Both the Ca2+-independent (basal) PKC activity representing the activation of PKC by the in vivo treatments and the Ca+2-dependent (total) PKC activity assayed in the presence of exogenous cofactors in vitro were determined. A comparison of the two activities demonstrated the strength and temporal status of PKC regulation by steroid hormones in vivo. P treatment resulted in a rapid increase in basal PKC activity in the VMN but not the POA. Estradiol benzoate priming augmented P-initiated increase in PKC basal activity in both the VMN and POA. These increases were inhibited by intracerebroventricular administration of a PKC inhibitor administered 30 min prior to P. The total PKC activity remained unchanged demonstrating maximal PKC activation within 30 min in the VMN. In contrast, P regulation in the POA significantly attenuated total PKC activity +/- estradiol benzoate priming. These rapid changes in P-initiated PKC activity were not due to changes in PKC protein levels or phosphorylation status.


Subject(s)
Brain/drug effects , Hypothalamus/drug effects , Progesterone/pharmacology , Protein Kinase C/metabolism , Animals , Brain/physiology , Calcium/metabolism , Calcium/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Female , Hypothalamus/enzymology , Hypothalamus/metabolism , Indoles/pharmacology , Maleimides/pharmacology , Ovariectomy , Phosphorylation/drug effects , Preoptic Area/drug effects , Preoptic Area/enzymology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
17.
Neuroreport ; 19(4): 431-5, 2008 Mar 05.
Article in English | MEDLINE | ID: mdl-18287941

ABSTRACT

In a previous study, we demonstrated that androgenic-anabolic steroids increased aromatase expression in the bed nucleus of stria terminalis and preoptic area in rat brain, as evaluated using autoradiography with [11C]vorozole, a potential positron emission tomography tracer for aromatase. In this study, we explored whether the increase in aromatase binding is mediated via androgen receptors and whether this increase occurs in neurons or glial cells. Rats were given nandrolone decanoate (15 mg/kg body weight once every 3 days) and flutamide (20 mg/kg/day) alone or in combination for 20 days. Results indicated a significant increase of [11C]vorozole binding by nandrolone decanoate in the bed nucleus of the stria terminalis and preoptic area, as in our previous study. Flutamide treatment, on the other hand, decreased [11C]vorozole binding in the bed nucleus of the stria terminalis, preoptic area, and medial amygdala. Immunohistochemical examination demonstrated that upregulation of aromatase expression occurred in neurons. Our findings suggest that aromatase is regulated through an androgen receptor-mediated system. This aromatase-specific tracer and the positron emission tomography technique could be useful for exploring the role of aromatase in anabolic androgenic steroids abusers.


Subject(s)
Androgens/pharmacology , Aromatase/metabolism , Brain/drug effects , Flutamide/pharmacology , Neurons/drug effects , Receptors, Androgen/drug effects , Amygdala/diagnostic imaging , Amygdala/drug effects , Amygdala/enzymology , Androgen Antagonists/pharmacology , Animals , Aromatase Inhibitors/metabolism , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/diagnostic imaging , Brain/enzymology , Brain Mapping , Carbon Radioisotopes , Male , Neurons/enzymology , Positron-Emission Tomography , Preoptic Area/diagnostic imaging , Preoptic Area/drug effects , Preoptic Area/enzymology , Rats , Rats, Sprague-Dawley , Receptors, Androgen/metabolism , Septal Nuclei/diagnostic imaging , Septal Nuclei/drug effects , Septal Nuclei/enzymology , Triazoles/metabolism , Triazoles/pharmacokinetics , Up-Regulation/drug effects , Up-Regulation/physiology
18.
J Comp Neurol ; 500(5): 894-907, 2007 Feb 10.
Article in English | MEDLINE | ID: mdl-17177261

ABSTRACT

In many vertebrate species the medial preoptic area projects to a premotor nucleus, the periaqueductal central gray (PAG). This connection plays an important role in the control of reproductive behavior. In male Japanese quail (Coturnix japonica) specifically, the medial preoptic nucleus (POM), where various types of sensory inputs converge, is a critical site for the activational action of testosterone on male sexual behavior. To activate male copulatory behavior, testosterone must be aromatized to estradiol within the POM and aromatase-immunoreactive cells in the POM are the main source of projections to the PAG. The POM-PAG connection is thus an important functional circuit integrating the sensory with premotor components of sexual behavior. Contrary to what is observed in males, testosterone does not activate male-typical copulatory behavior in females and we investigated here via retrograde tracing methods whether this behavioral sexual difference is associated with a sex difference in connectivity between POM and PAG. Fluorescent microspheres were injected in the PAG of male and female quail and retrogradely labeled fluorescent cells counted in four fields of the POM in sections that had been immunolabeled for aromatase. Males had more aromatase-immunoreactive neurons projecting to the PAG than females and this difference was most prominent in the caudolateral part of the nucleus that has been specifically implicated in the control of male copulatory behavior. These data therefore support the hypothesis that sex differences in POM-PAG connectivity are causally linked to the sex difference in the behavioral response to testosterone.


Subject(s)
Copulation/physiology , Neural Pathways/enzymology , Periaqueductal Gray/cytology , Preoptic Area/enzymology , Analysis of Variance , Animals , Coturnix , Female , Male , Neural Pathways/cytology , Periaqueductal Gray/metabolism , Preoptic Area/cytology , Sex Characteristics , Testosterone/metabolism
19.
J Neuroendocrinol ; 19(2): 139-41, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17214876

ABSTRACT

There are apparently normal male rats that fail to initiate copulation; these animals are called non-copulating (NC) males. Several research groups have demonstrated that conversion of testosterone to oestradiol (aromatisation) in specific brain areas known to be involved in the control of masculine sexual behaviour is fundamental in the control of masculine sexual behaviour. The aim of the present study was to test the hypothesis that the concentration of aromatase activity (AA) in the brain is lower in NC males than in copulating males (C). We quantified AA in several brain nuclei and also evaluated whether NC rats have altered concentrations of testosterone in their plasma. We found that AA was reduced in the medial preoptic nuclei (MPN) of NC male rats vs C males. In addition, NC and C male rats had similar plasma levels of testosterone. These data suggest that reduced levels of AA in the MPN could be a crucial factor associated with lack of male coital behaviour in rats.


Subject(s)
Aromatase/metabolism , Copulation/physiology , Neurons/enzymology , Preoptic Area/enzymology , Animals , Male , Preoptic Area/cytology , Rats , Sex Factors , Testosterone/blood
20.
J Neuroendocrinol ; 19(10): 767-72, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17850458

ABSTRACT

Steroid hormones act on developing neural circuits that regulate the hypothalamic-pituitary-gonadal axis and are involved in hormone-sensitive behaviours. To test the hypothesis that developmental exposure to oestradiol (E(2)) organises the quantity of adult oestrogen receptors (ERalpha and ERbeta), we used male mice with a targeted mutation of the aromatase enzyme gene (ArKO) and their wild-type (WT) littermates. These mice are unable to aromatise testosterone to E(2), but still express both ERalpha and beta. To evaluate adult responsiveness to E(2), gonadectomised males were implanted with Silastic capsules containing E(2), or an empty implant, 5 days prior to sacrifice. Immunoreactivity for ERalpha and ERbeta was quantified in the caudal ventromedial nucleus (VMN) and the medial preoptic area (POA). Regardless of genotype, adult treatment with E(2) reduced ERalpha-immunoreactive (ir) and ERbeta-ir cell numbers in the POA, as well as ERbeta-ir, but not ERalpha-ir, cell numbers in the VMN. Genotype, and thus endogenous exposure to E(2), produced opposite effects on ER expression in the two brain areas. In the VMN, ArKO males had more ERalpha-ir and ERbeta-ir cells than did WT males. In the POA, ArKO males had fewer ERalpha-ir and ERbeta-ir cells than did WT males. Thus, numbers of immunoreactive neurones containing both ERs in the adult ArKO male were enhanced in the POA, but decreased in the VMN, and most likely these patterns were established during the developmental critical period. Furthermore, although both ERalpha and beta-ir cell numbers are altered by the disruption of the aromatase gene, ERbeta is altered in a more robust and region-specific manner.


Subject(s)
Estradiol/physiology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Preoptic Area/metabolism , Sex Differentiation/physiology , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Aromatase/deficiency , Aromatase/metabolism , Critical Period, Psychological , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Preoptic Area/enzymology , Preoptic Area/growth & development , Tissue Distribution , Ventromedial Hypothalamic Nucleus/enzymology , Ventromedial Hypothalamic Nucleus/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL