scispace - formally typeset
Search or ask a question

Showing papers on "Hypothalamus published in 1995"


Journal ArticleDOI
05 Oct 1995-Nature
TL;DR: This study shows that D2 receptors have a key role in the dopaminergic control of nervous function, using homologous recombination to generate D2-receptor-deficient mice, which have therapeutic potential as a model for investigating and correcting dysfunctions of the dopamine system.
Abstract: Dopaminergic neuronal pathways arise from mesencephalic nuclei and project axons to the striatum, cortex, limbic system and hypothalamus. Through these pathways dopamine affects many physiological functions, such as the control of coordinated movement and hormone secretion. Here we have studied the physiological involvement of the dopamine D2 receptors in dopaminergic transmission, using homologous recombination to generate D2-receptor-deficient mice. Absence of D2 receptors leads to animals that are akinetic and bradykinetic in behavioural tests, and which show significantly reduced spontaneous movements. This phenotype presents analogies with symptoms characteristic of Parkinson's disease. Our study shows that D2 receptors have a key role in the dopaminergic control of nervous function. These mice have therapeutic potential as a model for investigating and correcting dysfunctions of the dopaminergic system.

658 citations


Journal ArticleDOI
TL;DR: This study used Western blot analysis and immunohistochemistry to describe the biochemical characterization and anatomical distribution of the second, mitogen‐inducible form of this enzyme, COX 2 in the rat brain.
Abstract: Considerable debate exists regarding the cellular source of prostaglandins in the mammalian central nervous system (CNS). At least two forms of prostaglandin endoperoxide synthase, or cyclooxygenase (COX), the principal enzyme in the biosynthesis of these mediators, are known to exist. Both forms have been identified in the CNS, but only the distribution of COX 1 has been mapped in detail. In this study, we used Western blot analysis and immunohistochemistry to describe the biochemical characterization and anatomical distribution of the second, mitogen-inducible form of this enzyme, COX 2 in the rat brain. COX 2-like immunoreactive (COX 2-ir) staining occurred in dendrites and cell bodies of neurons, structures that are typically postsynaptic. It was noted in distinct portions of specific cortical laminae and subcortical nuclei. The distribution in the CNS was quite different from COX 1. COX 2-ir neurons were primarily observed in the cortex and allocortical structures, such as the hippocampal formation and amygdala. Within the amygdala, neurons were primarily observed in the caudal and posterior part of the deep and cortical nuclei. In the diencephalon, COX 2-ir cells were also observed in the paraventricular nucleus of the hypothalamus and in the nuclei of the anteroventral region surrounding the third ventricle, including the vascular organ of the lamina terminalis. COX 2-ir neurons were also observed in the subparafascicular nucleus, the medial zona incerta, and pretectal area. In the brainstem, COX 2-ir neurons were observed in the dorsal raphe nucleus, the nucleus of the brachium of the inferior colliculus, and in the region of the subcoeruleus. The distribution of COX 2-ir neurons in the CNS suggests that COX 2 may be involved in processing and integration of visceral and special sensory input and in elaboration of the autonomic, endocrine, and behavioral responses.

541 citations


Journal ArticleDOI
TL;DR: Maladaptive neuroendocrine responses, i.e., dysregulation of the stress system, may lead to disturbances in growth and development, and cause psychiatric, endocrine/metabolic, and/or autoimmune diseases or vulnerability to such diseases.
Abstract: The human organism is in a state of dynamic equilibrium, homeostasis. The stress system is activated when homeostasis is challenged by extrinsic or intrinsic forces, the stressors. This system, whose central component is the central nervous system (CNS) and includes corticotropin-releasing hormone (CRH) and noradrenergic neurons, respectively, in the hypothalamus and the brain stem, has as its peripheral limbs the hypothalamic-pituitary-adrenal (HPA) axis and the autonomic (sympathetic) nervous system. Normal development and preservation of life and species are dependent on a normally functioning stress system. Maladaptive neuroendocrine responses, i.e., dysregulation of the stress system, may lead to disturbances in growth and development, and cause psychiatric, endocrine/metabolic, and/or autoimmune diseases or vulnerability to such diseases.

536 citations


Journal ArticleDOI
02 Feb 1995-Nature
TL;DR: It is reported here that corticotropin-releasing hormone-deficient mice reveal a fetal glucocorticoid requirement for lung maturation, suggesting that the major role of glucocORTicoid is during fetal rather than postnatal life.
Abstract: The body responds to stress by activation of the hypothalamic-pituitary-adrenal (HPA) axis and release of glucocorticoids. Glucocorticoid production in the adult regulates carbohydrate and amino-acid metabolism, maintains blood pressure, and restrains the inflammatory response. In the fetus, exogenous glucocorticoids accelerate maturation of lung and gastrointestinal enzyme systems and promote hepatic glycogen deposition. Corticotropin-releasing hormone (CRH), a 41-amino-acid neuropeptide produced in the paraventricular nucleus of the hypothalamus and many regions of the cerebral cortex, has been implicated in both the HPA axis and behavioural responses to stress. To define the importance of CRH in the response of the HPA axis to stress and fetal development, we have constructed a mammalian model of CRH deficiency by targeted mutation in embryonic stem (ES) cells. We report here that corticotropin-releasing hormone-deficient mice reveal a fetal glucocorticoid requirement for lung maturation. Postnatally, despite marked glucocorticoid deficiency, these mice exhibit normal growth, fertility and longevity, suggesting that the major role of glucocorticoid is during fetal rather than postnatal life.

510 citations


Journal ArticleDOI
TL;DR: The biochemical data support the idea that the central GLP‐1 receptor resembles the peripheral GLP-1 receptor, and the presence of GLp‐1 binding sites in the circumventricular organs suggests that these may be receptors which act as the target for both peripheral blood‐borne GLP­1 and GLP•1 in the nervous system.
Abstract: The distribution and biochemical properties of glucagon-like peptide (GLP)-1(7-36) amide (GLP-1) binding sites in the rat brain were investigated. By receptor autoradiography of tissue sections, the highest densities of [125I]GLP-1 binding sites were identified in the lateral septum, the subfornical organ (SFO), the thalamus, the hypothalamus, the interpenduncular nucleus, the posterodorsal tegmental nucleus, the area postrema (AP), the inferior olive and the nucleus of the solitary tract (NTS). Binding studies with [125I][Tyr39] exendin-4, a GLP-1 receptor agonist, showed an identical distribution pattern of binding sites. Binding specificity and affinity was investigated using sections of the brainstem containing the NTS. Binding of [125I]GLP-1 to the NTS was inhibited concentration-dependently by unlabelled GLP-1 and [Tyr39]exendin-4 with KI values of 3.5 and 9.4 nM respectively. Cross-linking of hypothalamic membranes with [125I]GLP-1 or [125I][Tyr39]exendin-4 identified a single ligand-binding protein complex with a molecular mass of 63,000 Da. The fact that no GLP-1 binding sites were detected in the cortex but that they were detected in the phylogenetically oldest parts of the brain emphasizes that GLP-1 may be involved in the regulation of vital functions. In conclusion, the biochemical data support the idea that the central GLP-1 receptor resembles the peripheral GLP-1 receptor. Furthermore, the presence of GLP-1 binding sites in the circumventricular organs suggests that these may be receptors which act as the target for both peripheral blood-borne GLP-1 and GLP-1 in the nervous system.

484 citations


Journal ArticleDOI
TL;DR: The common expression in both species of an IL‐1R in non‐neuronal elements highlights the possibility thatIL‐1‐mediated activation of CRF neurons may result from cytokine‐receptor interaction at vascular, and/or other barrier‐related, sites to trigger release of secondary signalling molecules in a position to interact with components of HPA control circuitry.
Abstract: Systemic interleukin-1 (IL-1) activates the hypothalamo-pituitary-adrenal (HPA) axis, an effect exerted through increased synthesis and secretion of corticotropin-releasing factor (CRF) by parvicellular neurosecretory neurons. The site(s) and mechanism(s) through which circulating IL-1 may access central systems governing HPA axis output remain obscure. To identify potential cellular targets for blood-borne IL-1, we analyzed the distribution of mRNA encoding the rat type 1 IL-1 receptor (IL-1R1) in rat brain. Regional ribonuclease protection assays detected a single protected fragment corresponding to the membrane-bound form of the IL-1R1 mRNA in all areas analyzed. In situ hybridization revealed labeling predominantly over barrier-related cells, including the leptomeninges, non-tanycytic portions of the ependyma, the choroid plexus, and vascular endothelium. Low to moderate levels of the IL-1R1 mRNA were detected in just a few neuronal cell groups, including the basolateral nucleus of the amygdala, the arcuate nucleus of the hypothalamus, the trigeminal and hypoglossal motor nuclei, and the area postrema. No specific labeling for IL-1R1 mRNA was detected over neurons that respond to intravenous IL-1 beta by induction of transcription factor Fos, including hypophysiotropic CRF cells and brainstem catecholamine neurons. Injection of IL-1 beta did, however, provoke induction of mRNA encoding the immediate-early gene, NGFI-B, but not c-fos, in two major loci of IL-1R1 expression, vascular endothelial cells, and the area postrema. Intravenous injection of IL-1 beta acutely down-regulated IL-1R1 mRNA in perivascular cells, but not in neuronal cell groups. These results suggest the parenchymal sites of IL-1R1 expression in rat to be distinct from those reported previously in mouse. The common expression in both species of an IL-1R in non-neuronal elements highlights the possibility that IL-1-mediated activation of CRF neurons may result from cytokine-receptor interaction at vascular, and/or other barrier-related, sites to trigger release of secondary signalling molecules in a position to interact with components of HPA control circuitry.

457 citations


Journal ArticleDOI
TL;DR: Paraventricular CRH neurons in Alzheimer's disease and depression are hyperactivated, and this hyperactivation may contribute to the etiology of these disorders.
Abstract: Objective: Greater activity of the hypothalamic-pituitary-adrenal (HPA) axis is associated with specific neurological and psychiatric disorders, including Alzheimer’s disease and depression. Hyperactivation ofparaventricular corticotropin-releasing hormone (CRH) neurons may form the basis of this increased activity of the HPA axis. Method: Activation of the CRH neurons was determined through measurement ofthe amount ofCRH-mRNA in the paraventricular nucleus by using quantitative, in situ hybridization histochemistry with systematically sampled frontal sections through the hypothalamus of routinely formalin-fixed and paraffinembedded autopsy brain material of 1 0 comparison subjects, 10 patients with Alzheimer’s disease, and seven depressed patients. Results: CRH-mRNA levels in the paraventricular nucleus ofAlzheimer’s patients were markedly higher than those ofcomparison subjects, whereas CRH-mRNA levels in the paraventricular nucleus ofdepressed patients were even higher than the levels ofAlzheimer’s patients. Conclusions: Paraventricular CRH neurons in Alzheimer’s disease and depression are hyperactivated, and this hyperactivation may contribute to the etiology of these disorders. (AmJ Psychiatry1995; 152:1372-1376)

403 citations


Journal ArticleDOI
TL;DR: The OXT neurons of the PVN seem to be good candidates for playing a physiological role in ingestive behavior as "satiety neurons" in the human hypothalamus.
Abstract: Animal experiments have shown that the parvocellular oxytocin (OXT) neurons of the hypothalamic paraventricular nucleus (PVN) inhibit food intake. In the present study, the PVN and its OXT neurons have been investigated in an extreme human eating disorder, i.e. the Prader-Willi syndrome (PWS). PWS patients are characterized by gross obesity, insatiable hunger, hypotonia, hypogonadism, and mental retardation. The PVN of 5 PWS patients (2 males and 3 females), varying in age between 22-64 yr, and 27 controls (14 males and 13 females) without any primary neurological or psychiatric diseases was morphometrically investigated after conventional staining with thionine and immunocytochemical staining for OXT and vasopressin (AVP). The thionine-stained volume of the PVN was 28% smaller in PWS patients (P = 0.028), and the total cell number was 38% lower (P = 0.009). The immunoreactivity for OXT and AVP was decreased in PWS patients, although the variability within the groups was high. A strong and highly significant decrease (42%; P = 0.016) was found in the number of OXT-expressing neurons of the PWS patients. The volume of the PVN-containing OXT-expressing neurons decreased by 54% (P = 0.028) in PWS. The number of AVP-expressing neurons in the PVN did not change significantly. The OXT neurons of the PVN seem to be good candidates for playing a physiological role in ingestive behavior as "satiety neurons" in the human hypothalamus.

387 citations


Journal ArticleDOI
01 Feb 1995-Diabetes
TL;DR: The results demonstrate that CNS infusion of insulin to diabetic rats reduces both hyperphagia and overexpression of hypothalamic NPY mRNA, which supports the hypothesis that a deficiency of insulin in the brain is an important cause of diabetic hyperphagy and that increased hypothalamicNPY gene expression contributes to this phenomenon.
Abstract: To test the hypothesis that diabetic hyperphagia results from insulin deficiency in the brain, diabetic rats (streptozotocin-induced) were given an intracerebroventricular (ICV) infusion of saline or insulin (at a dose that did not affect plasma glucose levels) for 6 days. Food and water intake were significantly increased in diabetic rats, but only food intake was affected by ICV insulin. Diabetic hyperphagia was reduced 58% by ICV insulin compared with ICV saline (P < 0.05) and was accompanied by a 69% increase in diabetes-induced weight loss (P < 0.05). To evaluate whether central nervous system (CNS) insulin deficiency affects expression of neuropeptides involved in food intake, in situ hybridization was done for neuropeptide Y (NPY), which stimulates feeding, in the hypothalamic arcuate nucleus and for cholecystokinin (CCK) and corticotropin-releasing hormone (CRH), which inhibit feeding, in the hypothalamic paraventricular nucleus. In diabetic rats, NPY mRNA hybridization increased 280% (P < 0.05), an effect reduced 40% by ICV insulin (P < 0.05). CCK mRNA hybridization increased 50% in diabetic rats (P < 0.05), a response reduced slightly by ICV insulin (P < 0.05), whereas CRH mRNA hybridization decreased 33% in diabetic rats (P < 0.05) and was unchanged by ICV insulin. The results demonstrate that CNS infusion of insulin to diabetic rats reduces both hyperphagia and overexpression of hypothalamic NPY mRNA. This observation supports the hypothesis that a deficiency of insulin in the brain is an important cause of diabetic hyperphagia and that increased hypothalamic NPY gene expression contributes to this phenomenon.

384 citations


Journal ArticleDOI
TL;DR: The results show that the anterior PVT is ideally situated to relay circadian timing information from theSCN to brain areas involved in visceral and motivational aspects of behavior and to provide feedback regulation of the SCN.
Abstract: The paraventricular nucleus of the thalamus (PVT) receives input from all major components of the circadian timing system, including the suprachiasmatic nucleus (SCN), the intergeniculate leaflet and the retina. For a better understanding of the role of this nucleus in circadian timing, we examined the distribution of its efferent projections using the anterograde tracer Phaseolus vulgaris leucoagglutinin (PHA-L). The efferent projections of the PVT are loosely organized along its dorsal-ventral and anterior-posterior axes. The anterior PVT sends projections to the SCN; the dorsomedial and ventromedial hypothalamic nuclei; the lateral septum; the bed nucleus of the stria terminalis; the central and basomedial amygdaloid nuclei; the anterior olfactory nucleus; the olfactory tubercle; the nucleus accumbens; the infralimbic, piriform, and perirhinal cortices; the ventral subiculum; and the endopiriform nucleus. A small PHA-L injection, restricted to the ventral portion of the anterior PVT, produces a similar pattern of labeling, except for a marked decrease in the number of labeled fibers in the hypothalamus, cortex, and lateral septum and an increase in labeling in the endopiriform nucleus and basolateral amygdaloid nucleus. The posterior PVT has a more limited efferent distribution than the anterior PVT, terminating in the anterior olfactory nucleus; the olfactory tubercle; the nucleus accumbens; and the central, basolateral, and basomedial nuclei of the amygdala. Our results show that the anterior PVT is ideally situated to relay circadian timing information from the SCN to brain areas involved in visceral and motivational aspects of behavior and to provide feedback regulation of the SCN.

382 citations


Journal ArticleDOI
TL;DR: The rhythm in melatonin production by the mammalian pineal gland is generated by the circadian timing system, and the components of that system are described.

Journal ArticleDOI
TL;DR: The data indicate that a CORT-mediated decrease in hippocampal and hypothalamic glucocorticoid receptor mRNA levels is not the only mechanism contributing to the maintenance of a robust HPA response after repeated stress and postulates that the relative shift from CRH to AVP in the PVN after repeated Stress is mediated by both a greater sensitivity of AVP to CORT negative feedback and Cort-independent mechanisms.
Abstract: Hypothalamic-pituitary-adrenal (HPA) responses remain intact or increase after chronic or repeated stress despite robust levels of circulating glucocorticoids that would be expected to restrain the responsiveness of the axis. The purpose of this study was to determine whether chronic stress altered corticosteroid receptor messenger RNA (mRNA) levels at any locus known to mediate glucocorticoid feedback on HPA function (i.e. hippocampus or hypothalamus), whether such effects were glucocorticoid dependent, and whether changes in corticosteroid receptor function could potentially contribute to the putative shift from corticotropin-releasing hormone (CRH) to arginine vasopressin (AVP) in the hypothalamic paraventricular nucleus (PVN) in the modulation of pituitary adrenal function occurring during chronic stress. We compared the stress responsiveness of sham-operated rats to that of adrenalectomized rats using a moderate dose of corticosterone (CORT) pellet replacement (ADX + CORT group). Acute immobilization caused a significant increase in CRH, but not AVP, mRNA levels in the parvocellular PVN in sham rats. The ADX + CORT group showed significantly greater increases in both CRH and AVP mRNA levels in the PVN compared to sham rats. These data indicate that PVN AVP mRNA levels are more sensitive to glucocorticoid negative feedback than are the levels of CRH mRNA. In repeated stress, the sham groups showed robust increases in PVN CRH and AVP mRNA levels despite high levels of plasma CORT. The rise in AVP mRNA levels was greater than that in CRH mRNA. Type II glucocorticoid receptor mRNA in the hippocampus and PVN was decreased in the repeatedly stressed sham group. These data suggest a decrease in the CORT negative feedback restraint of PVN CRH and AVP mRNA levels repeated stress and a persistence of relatively greater responsiveness of AVP mRNA levels to CORT negative feedback. After repeated stress in ADX+CORT rats, both PVN CRH and AVP mRNA levels showed robust responses, with a relatively greater increase in AVP mRNA. These data indicate that a CORT-mediated decrease in hippocampal and hypothalamic glucocorticoid receptor mRNA levels is not the only mechanism contributing to the maintenance of a robust HPA response after repeated stress. Similarly, we postulate that the relative shift from CRH to AVP in the PVN after repeated stress is mediated by both a greater sensitivity of AVP to CORT negative feedback and CORT-independent mechanisms.

Journal ArticleDOI
TL;DR: Exposure of animals to immobilization (IMMO) markedly and rapidly increases rates of synthesis, release, and metabolism of norepinephrine in all the brain areas mentioned above and supports previous suggestions that in the PVN NE stimulates release of corticotropin-releasing hormone (CRH).

Journal ArticleDOI
TL;DR: In this paper, double immunostaining of the pituitary for Ad4BP and trophic peptide hor- mones, FSH, TSH, and ACTH, indicated a re-stricted localization of the transcription factor to the gonadotroph.
Abstract: Ad4BP (or SF-1) has been iden- tified as a transcription factor which regulates all the steroidogenic P450 genes in the peripheral or- gans, and is encoded by the mammalian homo- logue of Drosophila FTZ-F1 gene. mRNA coding for Ad4BP was detected in the hypothalamus and pituitary of rats by RT-PCR. Immunohistochemi- cal analyses using an antiserum to Ad4BP in the brain and pituitary revealed that the transcrip- tion factor is expressed in nuclei of the dorso- medial part of the ventromedial hypothalamus (dmVMH) and in some subpopulation of the ade- nohypophysial cells. Double immunostaining of the pituitary for Ad4BP and trophic peptide hor- mones, FSH, TSH, and ACTH, indicated a re- stricted localization of Ad4BP to the gonadotroph. Disruption of the mouse Ftz-FI gene was clarified to induce severe defects in the organization of the dmVMH and the function of the pituitary gona- dotroph. However, some of the dm VMH neurons and pituitary gonadotrophs persisted, which pro- vided a sharp contrast to complete agenesis of the peripheral steroidogenic tissues (adrenal and go- nads) in the mutant mouse. Additional abnormal- ities were seen in the ventrolateral part of VMH and dorsomedial hypothalamic nucleus, both of which do not express Ad4BP but have strong reciprocal fiber-connections with the dmVMH. Aromatase P450-containing cells in the medial preoptico-amygdaloid region, which were devoid of Ad4BP, persisted even in the brain of the gene disrupted mice. The present results clearly showed that the hypothalamic and pituitary Ad4BPs are essential to normal development of the functional VMH and gonadotroph through some mechanism distinct from that in the periph-

Journal ArticleDOI
TL;DR: The aim of this study was to examine the afferents to the rat locus coeruleus by means of retrograde and anterograde tracing experiments using cholera-toxin B subunit and phaseolus leucoagglutinin to indicate that the area surrounding the locus coercedus is divided into individual nuclei with distinctAfferents.

Journal ArticleDOI
TL;DR: Results of this study demonstrate both similarities and differences in brain activation patterns following these two social behaviors and suggest that mating and agonistic behavior activate distinct neural circuits.

Journal ArticleDOI
TL;DR: Both neuronal and endocrine components of the hypothalamic-pituitary axis are critically dependent on the action of specific POU domain factors at a penultimate step in the sequential events that underlie the appearance of mature cellular phenotypes.
Abstract: Neurons comprising the endocrine hypothalamus are disposed in several nuclei that develop in tandem with their ultimate target the pituitary gland, and arise from a primordium in which three related class III POU domain factors, Brn-2, Brn-4, and Brn-1, are initially coexpressed. Subsequently, these factors exhibit stratified patterns of ontogenic expression, correlating with the appearance of distinct neuropeptides that define three major endocrine hypothalamic cell types. Strikingly, deletion of the Brn-2 genomic locus results in loss of endocrine hypothalamic nuclei and the posterior pituitary gland. Lack of Brn-2 does not affect initial hypothalamic developmental events, but instead results in a failure of differentiation to mature neurosecretory neurons of the paraventricular and supraoptic nuclei, characterized by an inability to activate genes encoding regulatory neuropeptides or to make correct axonal projections, with subsequent loss of these neurons. Thus, both neuronal and endocrine components of the hypothalamic-pituitary axis are critically dependent on the action of specific POU domain factors at a penultimate step in the sequential events that underlie the appearance of mature cellular phenotypes.

Journal ArticleDOI
TL;DR: The physiological importance of EAAs in the control of LH surge expression is evidenced by the findings that the steroid-induced LH surge in ovariectomized animals and the preovulatory LH Surge in cycling animals and in PMSG-primed animals is blocked by treatment with specific NMDA receptor antagonists, or non-NMDA receptors antagonists.
Abstract: Excitatory amino acids (EAAs), such as glutamate and aspartate, are found in large concentrations in presynaptic boutons of a variety of important hypothalamic nuclei, including the arcuate nucleus, supraoptic nucleus, suprachiasmatic nucleus, paraventricular nucleus, organum vasculosa of the lamina terminalis (OVLT) and preoptic area (POA). Likewise, the different ionotropic/metabotropic EAA receptor subtypes are found in the same regions of the hypothalamus although there are differences in their individual patterns of localization. Furthermore, there is evidence supporting the presence of ionotropic N-methyl-D-aspartate (NMDA) receptors and non-NMDA (kainate and AMPA) receptors in the anterior lobe, intermediate lobe and posterior lobe of the pituitary. The majority of work to date has focused on the role of EAAs in the control of LH secretion. Administration of glutamate, NMDA, kainate or AMPA leads to rapid LH release mediated through the stimulation of hypothalamic GnRH release. The major site of NMDA action appears to be the OVLT/preoptic area--where GnRH cell bodies reside, whereas AMPA and kainate have been suggested to act primarily at the arcuate nucleus/median eminence--the site of GnRH nerve terminals. There is evidence that some of the effects of glutamate on GnRH release may involve activation of the novel neurotransmitter nitric oxide and possibly catecholamines. The physiological importance of EAAs in the control of LH surge expression is evidenced by the findings that the steroid-induced LH surge in ovariectomized animals and the preovulatory LH surge in cycling animals and in PMSG-primed animals is blocked by treatment with specific NMDA receptor antagonists, or non-NMDA receptor antagonists. EAAs also appear to be important in regulating the normal pulsatile pattern of LH release as evidenced by the finding that both the NMDA antagonist, AP5, and the AMPA/kainate antagonist, DNQX, lower mean LH levels, LH pulse amplitude and LH pulse frequency in the adult ovariectomized rat. A role for NMDA receptors in the achievement of puberty has been suggested since activation of NMDA receptors has been shown to advance the time of vaginal opening in the immature female rat, while kainate and DNQX were without effect. Steroids have been reported not to affect NMDA receptor binding in the hypothalamus; however, steroids appear to up-regulate AMPA receptor GluR1 subunit levels and non-NMDA receptor binding in the hypothalamus. Steroids also increase the release rates of glutamate and aspartate in the POA during the steroid-induced LH surge in the ovariectomized adult rat.(ABSTRACT TRUNCATED AT 400 WORDS)

Journal ArticleDOI
TL;DR: Investigation of the long-term consequences of neonatal endotoxin exposure on hypothalamic-pituitary-adrenal axis (HPA) function in Long-Evans rats found that exposure to gram-negative LPS in early life can alter the development of neural systems which govern endocrine responses to stress and may thereby predispose individuals to stress-related pathology.
Abstract: The long-term consequences of neonatal endotoxin exposure on hypothalamic-pituitary-adrenal axis (HPA) function were assessed in adult female and male Long-Evans rats. At 3 and 5 d of age, pups were administered endotoxin (Salmonella enteritidis, 0.05 mg/kg, i.p.) at a dose that provokes a rapid and sustained physiological response, but with no mortality. As adults, neonatally endotoxin-treated animals exhibited significantly greater adrenocorticotrophic hormone (ACTH) and corticosterone responses to restraint stress than controls. In addition, dexamethasone pretreatment was less effective in suppressing ACTH responses to restraint stress in endotoxin-treated animals than in controls, suggesting decreased negative-feedback sensitivity to glucocorticoids. Neonatal endotoxin treatment elevated resting-state median eminence levels of corticotropin-releasing hormone (CRH) and arginine vasopressin in adult male animals, and arginine vasopressin in both adult males and females. Neonatal exposure to endotoxin also increased CRH mRNA expression in the paraventricular nucleus of the hypothalamus of adult males, with no difference in females. Finally, glucocorticoid receptor density was reduced across a wide range of brain regions in the neonatal endotoxin-treated, adult animals. These data illustrate the interactive nature of immune and endocrine systems during development. It appears that endotoxin exposure during critical stages of development decreases glucocorticoid negative-feedback inhibition of ACTH secretagogue synthesis, thus increasing HPA responsiveness to stress. The implication of these findings is that exposure to gram-negative LPS in early life can alter the development of neural systems which govern endocrine responses to stress and may thereby predispose individuals to stress-related pathology.

Journal ArticleDOI
TL;DR: The highly specific anatomical distribution of immunoreactive FGF-2 in neuronal and non-neuronal brain cells, supports the notion that it plays a multifunctional role in the CNS under normal physiology.

Journal ArticleDOI
TL;DR: It is postulated that the CRH and ACTH responses to a low dose of endotoxin are mediated by vagal afferents, and the responses to the high dose ofascular endotoxin involve additional neuronal or humoral pathways.
Abstract: In order to assess the possibility that endotoxin-induced activation of the hypothalamus-pituitary-adrenal (HPA) axis is mediated by vagal afferents, we studied the effects of transection of the vagal nerves on endotoxin-induced Fos expression in hypothalamic corticotropin-releasing hormone (CRH) neurons and plasma ACTH and corticosterone responses. Groups of rats were subjected to sham surgery, complete subdiaphragmatic vagotomy (SVGX), or selective transection of the hepatic branch (HVGX). Two weeks after surgery, endotoxin or saline was injected i.p. and rats were sacrificed by decapitation two hours later. SVGX blocked or attenuated the ACTH response to 20 and 250 micrograms/kg endotoxin, respectively. HVGX did not suppress the ACTH response to either endotoxin dose. In addition, corticosterone responses were not affected by SVGX or HVGX. The endotoxin-induced Fos expression in CRH neurons was suppressed in SVGX, but not in HVGX animals. These observations lead us to postulate that the CRH and ACTH responses to a low dose of endotoxin are mediated by vagal afferents. The responses to a high dose of endotoxin involve additional neuronal or humoral pathways.

Journal ArticleDOI
TL;DR: The results strongly suggest that Brn-2 plays an essential role in the determination and development of the PVN and SO neuronal lineages in the hypothalamus.
Abstract: We generated mice carrying a loss-of-function mutation in Brn-2, a gene encoding a nervous system specific POU transcription factor, by gene targeting in embryonic stem cells. In homozygous mutant embryos, migratory precursor cells for neurons of the paraventricular nuclei (PVN) and the supraoptic nuclei (SO) of the hypothalamus die at approximately E12.5. All homozygous mutants suffered mortality within 10 days after birth, possibly because of a complete deficiency of these neurons in the hypothalamus. Although neither developmental nor histological abnormalities were observed in heterozygous mice, the levels of expression of vasopressin and oxytocin in the hypothalamus of these animals were half these of wild-type mice. These results strongly suggest that Brn-2 plays an essential role in the determination and development of the PVN and SO neuronal lineages in the hypothalamus.

Journal ArticleDOI
TL;DR: It is suggested that IS enhances biologically active IL-1 in the hypothalamus, and that hypothalamic IL- 1 plays a role in the regulation of IS-induced responses including elevated monoamine release in theothalamus and activation of the hypothalamo-pituitary-adrenal axis.
Abstract: We investigated whether interleukin-1 (IL-1) activity in the rat hypothalamus was increased by immobilization stress (IS), and whether pretreatment with an interleukin-1 receptor antagonist (IL-1Ra) is capable of inhibiting IS-induced elevations of hypothalamic norepinephrine (NE), dopamine (DA), and serotonin (5-HT) and the levels of their metabolites as well as of plasma adrenocorticotropic hormone (ACTH). IL-1 activity was estimated with a bioassay using mouse thymocyte proliferation in the presence of concanavalin A. IL-1Ra was administered directly into the anterior hypothalamus, and monoamines were determined using a microdialysis technique and an HPLC system. First, we found that levels of IL-1 activity in the rat hypothalamus reached a maximum at 60 min after starting IS. Second, IL-1Ra (2 micrograms) significantly inhibited IS-induced increases in hypothalamic NE, DA, and 5-HT levels as well as the levels of their metabolites. In addition, IL-1Ra (2 micrograms) also inhibited the IS-induced elevation of plasma ACTH levels. Third, timing effects of IL-1Ra administration on the IS-induced monoamines or ACTH responses were examined. IL-1Ra (2 micrograms) administered at 5 or 60 min before the start of IS, but not at 5 or 60 min after IS had been started, exerted inhibitory effects on these responses, indicating that the effects of IL-1 occurred within 5 min after the initiation of IS. In summary, these results suggest that IS enhances biologically active IL-1 in the hypothalamus, and that hypothalamic IL-1 plays a role in the regulation of IS-induced responses including elevated monoamine release in the hypothalamus and activation of the hypothalamo-pituitary-adrenal axis. Moreover, since 5 min is too short a time for IS to induce production of IL-1, IS may augment the effects of preexisting IL-1 in the hypothalamus.

Journal ArticleDOI
TL;DR: The findings demonstrate that the systemic administration of nitroglycerin is capable of activating a spectrum of functionally diverse brain regions, which includes areas involved in reflex adjustments to nitrogoglyin-induced hypotension, areas involved with sensory nociceptive perception and areas associated with integrative regulation of autonomic, behavioral and neuroendocrine functions.

Journal ArticleDOI
TL;DR: The results suggest that BDNF is a stress-responsive intercellular messenger that may be an important component of the stress response of the hypothalamic-pituitary-adrenal axis.
Abstract: Brain-derived neurotropic factor (BDNF) is a member of the nerve growth factor family that is important for neuronal survival and plasticity. We recently demonstrated that stress decreases BDNF messenger RNA (mRNA) levels in the hippocampus, which raises the possibility that BDNF may play a role in regulation of the hypothalamic-pituitary-adrenal axis. The purpose of this study was to determine whether BDNF expression is present and influenced by stress in other brain areas relevant to control of the hypothalamic-pituitary-adrenal axis. Using in situ hybridization, we found that BDNF mRNA is present in the parvocellular portion of the hypothalamic paraventricular nucleus (PVN), the lateral hypothalamus, and the anterior and neurointermediate lobes of the pituitary in rats. Acute (2-h) or repeated immobilization stress increased BDNF mRNA in all of these areas. This was in distinct contrast to stress-induced decreases in extrahypothalamic areas, including the basolateral amygdala, claustrum, and cingulate cortex as well as the hippocampus. BDNF was expressed in both CRF and TRH neurons in the PVN. Reducing glucocorticoid or thyroid negative feedback increased BDNF mRNA in the PVN and anterior pituitary, but not in the neurointermediate lobe. These results suggest that BDNF is a stress-responsive intercellular messenger that may be an important component of the stress response.

Journal ArticleDOI
TL;DR: It is suggested that high concentrations of CORT or CRH synergistically decrease CRH-R messenger RNA (mRNA) levels in the AP, and that at least high CORT has an inhibitory effect on PVN CRh-R mRNA levels.
Abstract: Glucocorticoids and stress are known to influence the synthesis of corticotropin-releasing hormone (CRH) at a variety of sites in brain, including the hypothalamus and amygdala. The recent cloning of the CRH receptor (CRH-R) enabled us to determine whether glucocorticoids or stress influenced CRH action via regulation of CRH-R. We, therefore, used in situ hybridization to measure CRH-R messenger RNA (mRNA) levels in the hypothalamic paraventricular nucleus (PVN), anterior pituitary (AP), amygdala, and bed nucleus of the stria terminalis (BNST) under several conditions. Systemic corticosterone (CORT) treatment, both daily injection (5 mg/rat.day) up to 14 days and pellet implant (200 mg) for 14 days, decreased CRH-R mRNA in the PVN and lateral and basolateral nucleus of the amygdala (BLA). Corticosterone injection (10 mg/rat.day, for 7 days) decreased CRH-R mRNA in the AP. Adrenalectomy also decreased CRH-R mRNA in the PVN and AP, but did not alter it in the BLA. In both sham and adrenalectomized rats with...

Journal ArticleDOI
TL;DR: The present study investigated the effect of intraperitoneal administration of endotoxin lipopolysaccharide (LPS) and immobilization stress on the genetic expression of corticotropin-releasing factor receptor (CRF-R) in the brains of conscious male Sprague-Dawley rats.
Abstract: The present study investigated the effect of intraperitoneal (i.p.) administration of endotoxin lipopolysaccharide (LPS) and immobilization stress on the genetic expression of corticotropin-releasing factor receptor (CRF-R) in the brains of conscious male Sprague-Dawley rats. One group of rats was killed at 1, 3, 6, 9, and 12 hr after a single intraperitoneal injection of either the LPS (250 micrograms/100 gm of body weight) or the vehicle solution; the other group was killed before, immediately after, 1.5, 3, 6, and 12 hr after a 90 min acute session of immobilization stress. Rats were deeply anesthetized and rapidly perfused with a solution of 4% paraformaldehyde-borax. Frozen brains were mounted on a microtome and cut from the olfactory bulb to the medulla in 30 microns coronal sections. mRNA encoding the rat CRF-R was assayed by in situ hybridization histochemistry using a 35S-labeled riboprobe, and CRF-R localization within CRF-immunoreactive neurons in the PVN was determined using a combination of immunocytochemistry and in situ hybridization techniques. Strong basal levels of CRF-R transcripts were observed in several regions of the brain (piriform cortex, medial and basolateral nuclei of the amygdala, red nucleus, pontine gray, cerebellum, laterodorsal tegmental nucleus, caudal division of the zona incerta, nucleus incertus, spinal and principal sensory nuclei of the trigeminal nerve, and various layers of the cortex). A low to moderate signal was also detected in multiple sites (medial septal nucleus, nucleus of the diagonal band, supraoptic nucleus, arcuate nucleus of the hypothalamus, interpeduncular nucleus, and nucleus prepositus). Whereas vehicle-treated and control rats displayed hardly detectable signals of CRF-R mRNA in the paraventricular nucleus (PVN), CRF-R gene transcription was highly stimulated by LPS administration and immobilization stress in this hypothalamic structure. Indeed, the CRF-R mRNA signal was positive in the dorsomedial parvocellular PVN 3 hr after LPS injection, strong and maximum in both parvo- and magno-PVN at 6 hr postinjection, and declined 9 and 12 hr after treatment. Similarly, 90 min and 3 hr after the immobilization session, mRNA encoding the CRF-R was highly expressed in the parvo-PVN and totally vanished 12 hr after the stress. A lower but significant increase in the CRF-R transcript signal was also observed in the supraoptic nucleus 6 hr after the LPS treatment.

Journal ArticleDOI
TL;DR: Results provide evidence that i.p. endotoxin administration can produce a strong activation of various IEG transcripts in selective structures of the brain, which could in turn play a determinant role in the integration of immune input to the central nervous system.
Abstract: The present study investigated the effect of the acute-phase response of a systemic immune activation on the transcription of various immediate early genes (IEGs) and neuropeptides in the brain of conscious rats. One, 3, 6, 9, and 12 h after a single intraperitoneal (i.p.) administration of either the immune activator lipopolysaccharide (LPS) or the vehicle solution, adult male rats were sacrificed and their brains cut in 30-μm coronal sections, mRNA encoding the IEGs c-fos and nerve growth factor inducible-B (NGFI-B), and neuropeptides corticotropin-releasing factor (CRF), oxytocin (OT), and vasopressin (AVP) were assayed by in situ hybridization histochemistry using a 35S-labeled riboprobes. The primary transcripts {heteronuclear (hn)RNA} for these neuropeptides were also detected using intronic probe technology, and colocalization of c-fos mRNA within CRF, AVP, and OT neurons was determined by means of a combination of immunocytochemistry and in situ hybridization techniques on same the brain sections. One h after LPS treatment, both c-fos and NGFI-B genes were expressed in the parvocellular division of the paraventricular nucleus (PVN) of the hypothalamus. The medial preoptic area/organum vasculosum of the lamina terminalis, the supraoptic nucleus (SON), the magnocellular division of the PVN, the arcuate nucleus/median eminence, the locus coeruleus, the nucleus of the solitary tract, and the area postrema also exhibited a strong signal for these two transcripts 3 h after endotoxin administration. A smaller but a significant c-fos expression was observed in various structures, including the dorsomedial hypothalamic area, the central nucleus of the amygdala, the ventral part of the tuberomammillary nucleus, the laterodorsal tegmental nucleus, the external lateral part of the parabrachial nucleus, the dorsal division of the ambiguus nucleus, and the lateral reticular nucleus of LPS-injected rats. The signal for c-fos and NGFI-B mRNA in most of these brain nuclei reached a maximum at 3 h postinjection, declined at 6 h, and vanished 9 to 12 h after LPS treatment. In the parvocellular nucleus of the PVN, c-fos was largely expressed in CRF-immunoreactive (ir) neurons, whereas in the magnocellular part of that nucleus and in the SON, this transcript was colocalized in numerous OT-ir and few AVP-ir neurons. Relative levels of CRF mRNA in the parvocellular PVN were also significantly increased 6 h following LPS, but endotoxin did not alter the genetic expression of this stress-related neuropeptide in other brain regions. This was confirmed by the CRF primary transcript (hnRNA), which was expressed only in the PVN but not in any other nucleus in the brains of LPS-treated rats. Although AVP and OT mRNA levels were not significantly altered in either the PVN or the SON of immune-challenged rats because of the high basal levels of each transcript in these nuclei, LPS caused significant induction of AVP hnRNA in the parvoPVN. These results provide evidence that i.p. endotoxin administration can produce a strong activation of various IEG transcripts in selective structures of the brain, which could in turn play a determinant role in the integration of immune input to the central nervous system. It is possible that the stimulation of neuropeptide biosynthesis in the hypothalamic nuclei involved in the control of the hypothalamic-pituitary adrenal axis during immune challenge is an important step in this integration.

Journal ArticleDOI
TL;DR: The neural pathways between the hippocampus, septum and amygdala and the hypothalamus in relation to adrenocortical activity and the differential role of the medial forebrain bundle as well as the effects of various hypothalamic deafferentation on the transmission of the neural impulses to the hypothalamate are described.

Journal ArticleDOI
TL;DR: Three radioimmunoassays specific for PACAP38, PACAP27 and PRP demonstrate that all three preproPACAP peptides are expressed in the rat hypothalamus, implying roles as hypothalamic regulatory peptides.