Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where E.R. de Kloet is active.

Publication


Featured researches published by E.R. de Kloet.


The Lancet | 1988

Localisation of 11β-hydroxysteroid dehydrogenase-tissue specific protector of the mineralocorticoid receptor

Christopher R. W. Edwards; D. Burt; M.A. Mcintyre; E.R. de Kloet; Paul M. Stewart; L. Brett; W. Sutanto; C. Monder

In vitro the mineralocorticoid receptor is non-specific and does not distinguish between aldosterone and cortisol. In vivo certain tissues with this receptor are aldosterone selective (eg, kidney and parotid) whereas others with the same receptor are not (eg, hippocampus and heart). Experiments in rats showed that 11 beta-hydroxysteroid dehydrogenase (which converts cortisol to cortisone in man and corticosterone to 11-dehydrocorticosterone in the rat) was much more highly concentrated in aldosterone-selective tissues than in non-selective tissues. The localisation in the selective tissues was such that the enzyme could act as a paracrine or possibly an autocrine mechanism protecting the receptor from exposure to corticosterone. Autoradiographic studies showed that protection is lost when the enzyme is inhibited; 3H-corticosterone and 3H-aldosterone were bound to similar sites. These findings seem to explain why sodium retention, hypokalaemia, and hypertension develop in subjects with congenital deficiency of 11 beta-OHSD and those in whom the enzyme has been inhibited by liquorice.


Psychoneuroendocrinology | 1987

FEEDBACK ACTION AND TONIC INFLUENCE OF CORTICOSTEROIDS ON BRAIN FUNCTION: A CONCEPT ARISING FROM THE HETEROGENEITY OF BRAIN RECEPTOR SYSTEMS

E.R. de Kloet; J.M.H.M. Reul

Two types of corticosteroid receptors can be distinguished in rat brain. The type 1 receptor resembles the kidney mineralocorticoid receptor and has two functional expressions in brain, i.e. type 1 corticosterone (CORT) preferring sites (CR) and mineralocorticoid receptors (MR). The type 2 receptor is similar to the liver glucocorticoid receptor (GR). CORT binds to both CR and GR. The localization, binding specificity, and capacity of the receptor systems have served as criteria to evaluate steroid dependent events in brain biochemistry and behaviour. The GR is widely distributed in neurons and glial cells, with the highest density in frontal brain regions. The GR becomes occupied concomitant with rising plasma CORT levels after stress and as part of the circadian rhythm. The GR mediates the feedback action of CORT on stress-activated brain processes. The CR has its predominant localization in neurons of the septo-hippocampal complex and has a ten-fold higher affinity for CORT than that of the GR. The CR is, at all times of intact adrenocortical secretion, 90% or more occupied by endogenous hormone. The CR mediates a tonic influence exerted with stringent specificity by CORT on hippocampus-associated functions, e.g. cognition, mood, and affect. CORT, via the CR, thus contributes to hippocampus function in interpretation of sensory information, leading to appropriate neuroendocrine and behavioural responses, which are themselves subsequently subject to feedback action via the GR. The MR mediates the mineralocorticoid effect on salt and water balance and its behavioural corollary of salt appetite. The anatomical localization of the MR system is as yet ill-defined, although functional studies suggest circumventricular organs as mineralocorticoid target sites. The CR and the MR have in common the high affinity for mineralocorticoids, but the CR is defined by its exclusive responsiveness to CORT as its agonist. The CR and MR probably represent the same chemical receptor modality (type 1), which is expressed differentially depending on the presence of extravascular corticosteroid binding globulin (CBG) in the vicinity of the receptor. GR capacity is subject to autoregulation. Chronic stress, senescence, and chronic CORT administration reduce GR number, with, as a consequence, a less efficient feedback signal. The CR number seems not to be under the control of corticosteroids, probably since the receptor sites are extensively occupied by endogenous hormones. The CR number displays a circadian rhythm and is reduced during senescence.(ABSTRACT TRUNCATED AT 400 WORDS)


Progress in Neurobiology | 1994

Mineralocorticoid and glucocorticoid receptors in the brain. Implications for ion permeability and transmitter systems

Marian Joëls; E.R. de Kloet

In this review we have argued that corticosteroid hormones represent an endocrine signal that can influence neuronal communication. The steroids bind to intracellular receptors in the brain, resulting in slow effects that involve gene transcription, but they may also evoke rapid effects via membrane receptors. The signal carried by the corticosteroids is therefore divergent with respect to the dimension of space and time. Within the rat brain, at least two intracellular receptor subtypes, i.e. MRs and GRs, bind corticosterone. The affinity, density and localization of the MRs is different from the GRs, although the actual properties may vary somewhat depending on the condition of the animal. In general, due to the difference in affinity, low corticosteroid levels result in a predominant MR occupation, while higher steroid levels additionally occupy GRs. Recent studies indicate that predominant MR occupation is important for the maintenance of ongoing transmission in certain brain regions and for neuroprotection. By contrast, additional GR occupation (for a limited period of time) results in an attenuation of local excitability; yet, prolonged exposure to high steroid levels may become an endangering condition for neurons. Since predominant MR occupation on the one hand and additional GR occupation on the other hand induce different cellular actions, the ratio of MR/GR occupation is an important factor determining the net effect of corticosteroid hormones in the brain. How coordinated MR- and GR-mediated effects control neuronal communication under various physiological and pathological conditions will be a challenge for future research.


European Journal of Neuroscience | 1993

Cellular Localization of Interleukin 6 mRNA and Interleukin 6 Receptor mRNA in Rat Brain

B. Schöbitz; E.R. de Kloet; Win Sutanto; Florian Holsboer

The distribution of interleukin 6 (IL‐6) mRNA and IL‐6 receptor (IL‐6R) mRNA in the brain of adult male rats was studied at the light microscope level by in situ hybridization histochemistry using 35S‐labelled oligonucleotides. The transcripts of both genes were localized in the pyramidal neurons and in the granular neurons of the hippocampus, in neurons of the habenular nucleus as well as in the dorsomedial and ventromedial hypothalamus, in the piriform cortex, in scattered neurons of the cortex and in granular cells of the cerebellum. The medial preoptic nucleus and the anterior tip of the lateral ventricle contained mRNA encoding IL‐6 and its receptor. Moreover, white matter areas, such as the internal capsule, which consist of only fibres and glial cells, were found to have autoradiographic signals above background. The mRNAs for IL‐6 and IL‐6R in hippocampus and cerebellum are not different, as shown by Northern blot analyses of RNA isolated from these tissues. We postulate that the cytokine IL‐6 is expressed constitutively in discrete regions of the CNS and that it is involved in the mechanisms coordinating metabolic, behavioural and neuroendocrine changes not only during illness but also under normal physiological conditions. Our results suggest that IL‐6 mRNA and IL‐6R mRNA are colocalized, thus supporting a role of the cytokine in autocrine and paracrine communication.


Journal of Neuroendocrinology | 2006

Do Corticosteroids Damage the Brain

J. Herbert; Ian M. Goodyer; Ashley B. Grossman; Michael H. Hastings; E.R. de Kloet; Stafford L. Lightman; S. J. Lupien; Benno Roozendaal; Jonathan R. Seckl

Corticosteroids are an essential component of the bodys homeostatic system. In common with other such systems, this implies that corticosteroid levels in blood and, more importantly, in the tissues remain within an optimal range. It also implies that this range may vary according to circumstance. Lack of corticosteroids, such as untreated Addisons disease, can be fatal in humans. In this review, we are principally concerned with excess or disturbed patterns of circulating corticosteroids in the longer or shorter term, and the effects they have on the brain.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Point mutation in the mouse glucocorticoid receptor preventing DNA binding impairs spatial memory

Oitzl; Holger M. Reichardt; Marian Joëls; E.R. de Kloet

Activation of central glucocorticoid receptors caused by the stress that is associated with a learning task facilitates storage of the acquired information. The molecular mechanism underlying this phenomenon is entirely unknown. Glucocorticoid receptors can influence transcription both through DNA binding-dependent and -independent mechanisms. To assess the importance of these two modes of action for spatial memory, we here used male mutant mice in which homodimerization and DNA binding of the glucocorticoid receptor is largely prevented (GRdim/dim) while protein–protein interactions still can take place. These mice showed a selective impairment of spatial memory in the water maze. Locomotion and anxiety-related parameters measured in an open field and a light/dark preference task were comparable for mutant and control mice. Mutant mice released more corticosterone than control mice under basal resting conditions and in response to swimming, which could have influenced memory processes of the mice. However, mimicking the task-related increase in corticosterone by supplementary injection of corticosterone (250 μg/kg, i.p.) in adrenalectomized mice, resulting in equal plasma corticosterone concentrations in both genotypes, improved spatial memory of control mice but had no effect on mutant mice. These findings suggest that task-related facilitating effects of corticosterone on spatial memory indeed depend on DNA binding of the glucocorticoid receptor rather than on protein–protein interactions of the receptor with other transcription factors. Although it cannot be excluded that both processes are involved in a coordinated way, interrupting the DNA-binding capacity of the receptor is sufficient to induce impairment.


The Journal of Steroid Biochemistry and Molecular Biology | 2002

Glucocorticoid receptor variants: clinical implications

R.H DeRijk; Marcel J. M. Schaaf; E.R. de Kloet

Following exposure to stress, cortisol is secreted from the adrenal cortex under the control of the hypothalamic-pituitary-adrenal axis (HPA-axis). Central in the regulation of the HPA-axis is a two tied corticosteroid-receptor system, comprised of high and low affinity receptors, the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR), respectively. In addition, these corticosteroid receptors mediate the effects of cortisol during stress on both central and peripheral targets. Cortisol modulates gene-expression of corticosteroid-responsive genes, with the effect lasting from hours to days. Mutations in the GR-gene are being associated with corticosteroid resistance and haematological malignancies, although these mutations are relatively rare and probably not a common cause of these diseases. However, several GR-gene variants and single nucleotide polymorphisms (SNP) in the GR-gene have been identified which are relatively common in the human population. The GRbeta-variant, for example, has been proposed to influence corticosteroid-sensitivity and most evidence has been derived from the immune system and in particular asthma. With respect to polymorphisms, a BclI restriction fragment polymorphism and a Asp363Ser have been described, which not only influence the regulation of the HPA-axis, but are also associated with changes in metabolism and cardiovascular control. These associations of a GR-gene polymorphism with metabolism and cardivascular control, and also with the regulation of the HPA-axis, indicates an important underlying role of cortisol in the etiology of these complex disorders. Therefore, we propose that a common underlying defect in these complex disorders is a disregulation of the HPA-axis, especially during stress. The clinical implication is that the regulation of the HPA-axis should be envisioned as a primary target of new drugs for the treatment of stress-related disorders.


Stress | 2000

Corticosterone Effects on BDNF Expression in the Hippocampus Implications for Memory Formation

Marcel J. M. Schaaf; E.R. de Kloet; Erno Vreugdenhil

The adrenal steroid corticosterone has profound effect on the structure and function of the hippocampus. Probably as a result of that, it modulates memory formation. In this review, the question is addressed if the corticosterone effects on memory processes are mediated by alterations in the expression of the neurotrophin Brain-Derived Neurotrophic Factor (BDNF) in the hippocampus. First, studies are described investigating the effect of corticosterone on BDNF expression in the rat hippocampus. It appears that corticosterone suppresses the BDNF expression at the mRNA and protein level in a subfield-specific way. Second, a model for the mechanism of action is proposed. In this model, activated mineralocorticoid and glucocorticoid receptors repress transcriptional activity of the BDNF promoter site-specifically via interaction with other transcription factors. Third, the implications for learning and memory are discussed. Studies show that during water maze training, corticosterone levels rise significantly, but the BDNF expression is not suppressed in any hippocampal subfield. Furthermore, high BDNF expression levels in specific subfields correlate with a good memory performance. Therefore, we suggest that the resistance of the hippocampal BDNF expression to suppression by corticosterone, as seen after water maze training, may contribute to an optimal memory performance.


Neuroendocrinology | 1972

Brain RNA and Hypophysectomy; A Topographical Study

W.H. Gispen; P. Schotman; E.R. de Kloet

The present study was designed to localize the effect of hypophysectomy on brain RNA content at a topographical level. For this purpose, a brain dissection method, based on morphological dif-ferences between various brain areas, was developed. In these areas, total cell RNA was measured and expressed as the RNA/DNA ratio. The highest ratio was found in the cortex cerebri (ca. 2.15) and hippocampus (2.21), whereas cerebellum (0.42) and bulbus olfactorius (0.95) showed the lowest values. Hypophysectomy reduced the RNA content mainly of brain-stem structures (ca.–24%), i.e., the thalamus, hypothalamus, mesencephalon, and medulla oblongata. Nevertheless, a small but significant reduction was found in the rostral cortex. In view of previous studies, it is suggested that this disturbed RNA metabolism in brain-stem structures could have a bearing on the impaired avoidance performance of hypophysectomized rats.


International Journal of Developmental Neuroscience | 2003

The postnatal development of the hypothalamic-pituitary-adrenal axis in the mouse.

Mathias V. Schmidt; Leo Enthoven; M. van der Mark; Seymour Levine; E.R. de Kloet; Melly S. Oitzl

The main characteristic of the postnatal development of the stress system in the rat is the so‐called stress hypo‐responsive period (SHRP). Lasting from postnatal day (pnd) 4 to pnd 14, this period is characterized by very low basal corticosterone levels and an inability of mild stressors to induce an enhanced ACTH and corticosterone release. During the last years, the mouse has become a generally used animal in stress research, also due to the wide availability of genetically modified mouse strains. However, very few data are available on the ontogeny of the stress system in the mouse. This study therefore describes the postnatal ontogeny of peripheral and central aspects of the hypothalamic–pituitary–adrenal (HPA) axis in the mouse. We measured ACTH and corticosterone in blood and CRH, urocortin 3 (UCN3), mineralocorticoid receptor (MR) and glucocorticoid receptor (GR) transcripts in the brain at postnatal days 1, 2, 4, 6, 9, 12, 14 and 16. Our results show that we can subdivide the postnatal development of the HPA axis in the mouse in two phases. The first phase corresponds to the SHRP in the rat and lasts from right after birth (pnd 1) until pnd 12. Basal corticosterone levels were low and novelty exposure did not enhance corticosterone or ACTH levels. This period is further characterized by a high expression of CRH in the paraventricular nucleus (PVN) of the hypothalamus. Expression levels of GR in the hippocampus and UCN3 in the perifornical area are low at birth but increase significantly during the SHRP, both reaching the highest expression level at pnd 12. In the second phase, the mice have developed past the SHRP and were now exhibiting enhanced corticosterone basal levels and a response of ACTH and corticosterone to mild novelty stress. CRH expression was decreased significantly, while expression of UCN3 and GR remained high, with a small decrease at pnd 16. The expression of MR in the hippocampus was very dynamic throughout the postnatal development of the HPA axis and changed in a time and subregion specific manner. These results demonstrate for the first time the correlation between the postnatal endocrine development of the mouse and gene expression changes of central regulators of HPA axis function.

Collaboration


Dive into the E.R. de Kloet's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erno Vreugdenhil

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. van Os

Maastricht University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge