• Non ci sono risultati.

Hormones, Stress and Depression Marianne B. Müller

N/A
N/A
Protected

Academic year: 2022

Condividi "Hormones, Stress and Depression Marianne B. Müller"

Copied!
10
0
0

Testo completo

(1)

Marianne B. Müller

1

and Florian Holsboer

1

Summary

Every disturbance of the body, either real or imagined, evokes a stress response.

Essential to this stress response is the activation of the hypothalamic-pituitary- adrenocortical (HPA) system, finally resulting in the release of glucocorticoid hormones from the adrenal cortex. Glucocorticoid hormones, in turn, feed back to this system by central activation of two types of corticosteroid receptors:

the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR).

Whereas a brief period of controllable stress, experienced with general arousal and excitement, can be a challenge and might thus be beneficial, chronically elevated levels of circulating corticosteroid hormones are believed to enhance vulnerability to a variety of diseases, including human affective disorders.

The cumulative evidence makes a strong case implicating corticosteroid receptor dysfunction in the pathogenesis of affective disorders. Corticosteroid receptor dysfunction is followed by changes in the sensitivity of the system to the inhibitory effects of glucocorticoids on the synthesis of CRH and vasopressin in hypothalamic neurons. Changes in CRH and vasopressin levels, in turn, determine the responsiveness of the axis to subsequent stressors: increased production of these neuropeptides leads to increased HPA responses to stress and might be associated with an enhanced anxiety state. Although definitive controlled trials remain to be conducted, there is evidence indicating that cortisol-lowering or corticosteroid receptor antagonist treatments, as well as CRH type 1 receptor antagonists, may be of clinical benefit in individuals with major depression. Therefore, a more detailed knowledge of GR and CRH receptor signalling pathways will ultimately lead to the development of novel neuropharmacological intervention strategies.

Kordon et al.

Hormones and the Brain

© Springer-Verlag Berlin Heidelberg 2005

1

Max Planck Institute of Psychiatry, Kraepelinstrasse 2–10, 80804 Munich, Germany

(2)

Introduction

The stress hormone (hypothalamic-pituitary-adrenocortical; HPA) system:

corticosteroid hormones are the main actors in restoring homeostasis

Every disturbance of the body, either real or imagined, either physical or psychological, evokes a stress response. This stress response involves a large number of mechanisms and processes which, all together, serve to restrain the body’s defense reactions to stress, so as to restore homeostasis and to facilitate adaptation (Selye 1946). A brief period of controllable stress, experienced with general arousal and excitement, can be a challenge and might thus be beneficial.

In contrast, lack of control and persistent uncertainty in prolonged stressful situations can lead to a chronic state of "distress," resulting in chronically elevated levels of circulating corticosteroid hormones ("stress hormones"), which is believed to enhance vulnerability to a variety of diseases. Chronic exposure to elevated corticosteroid levels has been associated with several pathophysiological conditions, such as dysfunction of the immune system, neurodegeneration in aging and human affective disorders (Chrousos and Gold 1992; Mc Ewen 2000).

Essential to the stress response are hypothalamic paraventricular neurons expressing corticotropin-releasing hormone (CRH) and other co-secretagogues, such as vasopressin. CRH is the primary hypothalamic hypophysiotropic factor regulating both basal and stress-induced release of pituitary corticotropin (ACTH; Vale et al. 1981; Owens and Nemeroff 1991; Aguilera 1998). CRH triggers the immediate release of ACTH from the anterior pituitary, which, in turn, potently stimulates the release of glucocorticoids from the zona fasciculata of the adrenal cortex.

Corticosteroids feed back to the HPA system via two major negative

feedback routes, one directly at the level of the paraventricular nucleus of the

hypothalamus, parvocellular part to control hypothalamic CRH expression

(Erkut et al. 1998) and at the level of the anterior pituitary. This immediate

negative feedback is a fundamental way in which the HPA system is restrained

during stress and activity, and this restraint of HPA activation by glucocorticoids

is rapid and profound (for review, see Herman and Cullinan 1997). In contrast,

induction of CRH expression by increasing glucocorticoid levels has been

described to occur at the level of the central amygdala and the bed nucleus of the

stria terminalis (BNST; for review, see Watts 1996; Schulkin et al. 1998). The latter

is derived embryologically from the amygdala and plays a fundamental role in

the regulation of the HPA system during stress. This “paradoxical’ elevation of

CRH gene expression by glucocorticoids in the central nucleus of the amygdala

and lateral BNST may underlie a number of functional as well as pathological

emotional states in which elevated circulating levels of glucocorticoids are

accompanied by increased anxiety, such as in human affective disorders (for

review, see Holsboer 1995, 2000).

(3)

In concert with other components of the stress hormone system, the action of corticosteroid hormones displays two modes of operation (for review, see de Kloet et al. 1998). On the one hand, corticosteroids maintain basal activity of the HPA system and control the sensitivity or threshold of the system’s response to stress. On the other hand, corticosteroid feedback helps to terminate stress-induced HPA system activation and to restrain the stress response.

Corticosteroid hormones, therefore, facilitate an individual‘s ability to cope with, adapt to and recover from stress.

Due to their lipophilic nature, glucocorticoid hormones are able to easily penetrate through cell membranes into every cell of the body. Physiological control of the access of endogenous glucocorticoids into the brain is an important issue, all the more so as both glucocorticoid overexposure and glucocorticoid deficiency have been shown to potentially impair and endanger neuronal integrity and function. Accordingly, the potentially disruptive effects of corticosteroids in control of brain function and behaviour have received much attention over the last years (Uno et al. 1989; Lucassen et al. 2001; Müller et al. 2001). In this context, the recent findings that endogenous corticosteroid hormones are substrates of the multidrug-resistance gene 1-type P-glycoproteins are of particular importance (Meijer et al. 2000; Karssen et al. 2001; Müller et al.

2003a): mdr 1-type P-glycoproteins are highly expressed at the blood-brain- barrier to protect the central nervous system against the entry and accumulation of a wide range of toxic xenobiotics and drugs by actively transporting them against a concentration gradient.

Glucocorticoid receptors (GRs) occur everywhere in the brain but are most abundant in hypothalamic CRH neurons and pituitary corticotropes.

Mineralocorticoid receptors (MRs), in contrast, are highly expressed in the hippocampus and, at lower expression levels, in hypothalamic sites involved in the regulation of salt appetite and autonomic outflow. Interestingly, the MRs and GRs are co-localized in those brain structures that are involved in the regulation of fear and anxiety, such as hippocampus, septum and amygdala.

The two corticosteroid receptor types differ not only in their neuroanatomical distribution but also in their affinity and binding capacity for corticosteroids:

the MR binds GCs with a t10-fold higher affinity than does the GR (Reul and

de Kloet 1985). These findings on corticosteroid receptor diversity led to the

working hypothesis that the tonic influences of corticosterone are exerted via

hippocampal MRs, whereas the additional occupancy of GRs with higher levels

of corticosterone, e.g., following stress, mediates feedback actions aimed to

restrain HPA system activation. Thus, the balance in MR- and GR-mediated

effects exerted by corticosterone seems to be critical for homeostatic control (de

Kloet et al. 1997,1998).

(4)

Impaired stress hormone regulation – a risk factor for depression?

Several research groups formulated a hypothesis relating aberrant stress hormone regulation to causality of depression (for review, see Holsboer 1999, 2000). The reason for HPA hyperactivity and, in particular, for enhanced synthesis and release of CRH in depression is not yet clear. Genetic and experience-related factors may interact to induce manifold changes in corticosteroid receptor signaling, finally resulting in hypersecretion of both CRH and vasopressin (Raadsheer et al. 1994; Purba et al. 1996; de Kloet et al. 1998; Müller et al. 2000).

A considerable amount of evidence has been accumulated suggesting that normalization of the HPA system might be the final step necessary for stable remission of the disease (Holsboer and Barden 1996; Zobel et al. 1999; Holsboer 2000). On the basis of these findings, it has been further hypothesized that antidepressants may act through normalization of HPA system function (for review, see Holsboer 2000).

Prominent HPA abnormalities among patients with major depression are increased numbers of ACTH and cortisol secretory pulses, which is also reflected in elevated urinary cortisol secretion rates (Rubin et al. 1987).

These studies were complemented by different neuroendocrine function tests, including the dexamethasone suppression test (DST) and the combined dexamethasone-CRH-challenge test (dex-CRH test). This dex-CRH-test, combining the DST with CRH stimulation, has been shown to be a sensitive test for detecting HPA dysregulation in patients with affective disorders. In fact, Heuser and co-workers concluded from their studies that the sensitivity of this combined dex-CRH test (i.e., the likelihood to differentiate between normal and pathological states) is above 80%, depending on age and gender (Heuser et al. 1994). Whereas CRH-elicited ACTH response is blunted in depressives, dexamethasone pretreatment produces the opposite effect and paradoxically enhances ACTH release following CRH. Similarly, CRH-induced cortisol release is much higher in dexamethasone-pretreated patients than following a challenge with CRH alone. Moreover, the combined dex-CRH test has proved to be particularly useful as a predictor of increased risk for relapse: in those patients where the neuroendocrine abnormality persists, the risk of relapse or resistance to treatment is much higher (Holsboer et al. 1987; Heuser et al. 1996;

Zobel et al. 1999).

Such a HPA disturbance in feedback control can be acquired as a result of stressful life experiences and can be compounded by age (Brunson et al.

2001), or it can be genetically predetermined at all levels involved in fine-tuned

neuroendocrine regulation. Depressive and anxiety disorders are likely to

have significant genetic components, as suggested by familial segregation of

these disorders and twin and adoption studies. It is possible, however, that the

genetic component is not an independent risk factor for the development of these

disorders but rather a vulnerability factor predisposing for the development of

affective disorders following stress exposure. Likewise, genetic factors may also

(5)

protect an individual from stress-related illness. Genetic and experience-related factors may interact to induce manifold changes in corticosteroid receptor signaling, finally resulting in hypersecretion of both CRH and AVP (Raadsheer et al. 1994; Purba et al. 1996; de Kloet et al. 1998; Müller et al. 2000). In this context, the data from the Munich Vulnerability Study are of particular interest (Holsboer et al. 1995; Modell et al. 1997, 1998). This study investigated whether the HPA feedback disturbance observed among patients with major depression is present in otherwise healthy individuals who are at high risk for psychiatric disorders because they have a first-degree relative with an affective illness.

The Munich Vulnerability Study revealed that subjects who never suffered from a psychiatric disorder, but belong to families with a considerably high genetic load for depression, may display abnormal responses to the combined dex-CRH-challenge test (Holsboer et al. 1995; Lauer et al. 1998; Modell et al.

1998). In the absence of any previous depression in these individuals, or other life events or lifestyles that would explain the functional corticosteroid receptor changes as an enduring adaptation to major stress, the findings from this study are best understood as indicating a genetically transmitted risk factor, possibly rendering these individuals susceptible to affective disorders. In a follow-up study, 14 of the initial 47 high-risk probands were re-examined about four years after the index investigation and showed surprisingly constant results in the combined dex-CRH challenge test, so that one of the requirements for a vulnerability marker is fulfilled (Modell et al. 1998).

Whereas genetic linkage studies so far reject the glucocorticoid receptor gene as a possible gene of inherited pathology (Detera-Wadleigh et al. 1992; Mirow et al. 1994; Morisette et al. 1999; Moutsatsou et al. 2000), the recent development of refined genetic technologies opens up the possibility for large-scale screening of single nucleotide polymorphisms (SNPs; Syvanen 2001; Cowan et al. 2002).

These SNPs are highly abundant and are estimated to occur at one out of every 1,000 bases in the human genome. Depending on where a SNP occurs, it might have different consequences at the phenotypic level. The reason for the current interest in SNPs is the hope that they could be used as markers to identify genes that predispose individuals to complex, polygenic disorders. A recent investigation provided evidence that carriers of a specific polymorphism (ER22/23EK) in the GR gene are less sensitive to the suppressive effects of low- dose dexamethasone (van Rossum et al. 2002) and are suspected to be relatively more resistant to the effects of GC, with respect to the sensitivity of adrenal feedback mechanisms, than are non-carriers. Considering the aforementioned implications of HPA system dysregulation for the development and maintenance of affective disorder, those functionally relevant polymorphisms of the GR, therefore, may alter an innate setpoint for susceptibility to stress-associated psychiatric disorders (DeRijk et al. 2002).

As outlined above, there is robust evidence for a hyperactivtity of the

HPA system in human affective disorders, and it has been proposed that

(6)

hypercortisolemia is in some way integral to the pathogenesis and maintenance of psychopathology and cognitive deficits in these diseases. It is, therefore, not surprising that research into novel therapeutic approaches to depression has focused on strategies designed to modulate the effects of hypercortisolemia and/or the mechanisms underlying this phenomenon (Murphy 1997; McQuade and Young 2000). Antiglucocorticoid drugs and approaches in the treatment of human affective disorder include the administration of steroid-synthesis inhibitors, such as ketoconazole, metyrapone and aminogluthetimide (e.g., Ravaris et al. 1988; Malison et al. 1999; Wolkowitz et al. 1999) or GR antagonists, such as mifepristone (Belanoff et al. 2002).

Pathological anxiety is a frequent concomitant of major depression.

Moreover, there is a longstanding debate about whether anxiety and depression constitute different aspects of the same disorder (“continuum hypothesis”) or distinct, yet overlapping, conditions. Anxiety-related behavior is one of the most important, highly conserved behaviors among all kind of species that can be reliably measured in specific experimental paradigms in rodents. Increased CRH neuronal activity has been hypothesized both in major depression and anxiety disorders. The crucial role of CRHR1 in mediating anxiety-related behavior was confirmed in transgenic mice with a functional null mutation of CRHR1 (Smith et al. 1998; Timpl et al. 1998). Most recently, using a brain area-specific conditional knockout mouse line, researchers showed that limbic Crhr1 neuronal circuitries mediate anxiety-related behavior, and that this anxiolytic effect is independent of HPA system activity (Müller et al. 2003b). These results, therefore, allow us to genetically dissect CRH/Crhr1 neuronal pathways modulating behavior from those regulating neuroendocrine function, underlining the importance of limbic CRH/Crhr1 neuronal pathways as a promising pharmacological target for the treatment of human affective disorders, such as major depression or the consequences of early-life stress (Holsboer 1999; Keck and Holsboer 2001).

One such selective CRHR1 antagonist, NBI-30775 (also referred to as R121919), was first shown to decrease anxiety- and fear-related behaviors in response to stress in rodents (Keck et al. 2001). In an open-label proof of concept study, 20 patients with major depression receiving NBI-30775 experienced significant reductions in depression and anxiety ratings, whereas neither basal nor CRH-stimulated HPA system activity was impaired (Zobel et al. 2000).

Besides pharmacological and other more traditional approaches to examine

stress-anxiety interactions in animal models, an alternative way is to directly

modify neurobiological systems involved in the stress response and to examine

the effects of these manipulations on anxiety-related behaviors. As mentioned

before, the primary candidates for these types of manipulations are the various

components of the HPA system (for review, see Müller et al. 2002; Müller and

Keck 2002). The ability to generate mice with specific mutations or complete

deletions of certain genes of interest through homologous recombination in

embryonic stem cells has facilitated the study of many biological processes, from

(7)

embryogenesis to animal behavior. The generation of genetically engineered mice (e.g., "conventional" and "conditional" knock-outs) has allowed us to specifically target individual genes involved in stress hormone regulation. The identification and detailed characterization of these molecular pathways will ultimately lead to the development of novel neuropharmacological intervention strategies.

References

Aguilera G (1998) Corticotropin releasing hormone, receptor regulation and the stress response. Trends Endocrinol Metab 9: 329-336.

Belanoff JK, Rothschild AJ, Cassidy F, De Battista C, Baulieu EE, Schold C, Schatzberg AF (2002) An open label trial of C-1073 (mifepristone) for psychotic major depression. Biol Psychiat 52: 386-392.

Brunson KL, Avishai-Eliner S, Hatalski CG, Baram TZ (2001) Neurobiology of the stress response early in life: evolution of a concept and the role of corticotropin-releasing hormone. Mol Psychiat 6: 647-656.

Chrousos PW, Gold PW (1992) The concepts of stress and stress system disorders. Overview of physical and behavioural homeostasis. JAMA 267: 1244-1252.

Cowan WM, Kopnisky KL, Hyman SE (2002) The human genome project and its impact on psychiatry. Ann Rev Neurosci 25: 1-50.

de Kloet ER, Vreugdenhil E, Oitzl MS, Joels M (1997) Glucocorticoid feedback resistance.

Trends Endocrinol Metab 8: 26-33.

de Kloet ER, Vreugdenhil E, Oitzl MS, Joels M (1998) Brain corticosteroid receptor balance in health and disease. Endocrine Rev 19: 269-301.

DeRijk RH, Schaaf M, de Kloet ER (2002) Glucocorticoid receptor variants: clinical implications. J Steroid Biochem Mol Biol 81: 103-122.

Detera-Wadleigh SD, Berrettini WH, Goldin LR, Martinez M, Hsieh WT, Hoehe MR, Encio IJ, Coffman D, Rollins DY, Muniec D (1992) A systematic search for a bipolar predisposing locus on chromosome 5. Neuropsychopharmacology 6: 219-229.

Erkut ZA, Pool C, Swaab DF (1998) Glucocorticoids suppress corticotropin-releasing hormone and vasopressin expression in human hypothalamic neurons. J Clin Endocrinol Metab 83:

2066-2073.

Herman JP, Cullinan WE (1997) Neurocircuitry of stress: central control of the hypothalamic- pituitary-adrenocortical axis. Trends Neurosci 20: 78-84.

Heuser I, Yassouridis A, Holsboer F (1994) The combined dexamethasone/CRH test: a refined laboratory test for psychiatric disorders. J Psychiatr Res 28: 341-356.

Heuser IJE, Schweiger U, Gotthardt U, Schmider J, Lammers CH, Dettling M, Yassouridis A, Holsboer F (1996) Pituitary-adrenal system regulation and psychopathology during amitriptyline treatment in elderly depressed patients and in normal control subjects. Am J Psychiat 153: 93-99.

Holsboer F (1995) Neuroendocrinology of mood disorders. In: Bloom FE, Kupfer DJ (eds) Psychopharmacology: The fourth generation of progress. Raven Press, New York, pp.

957-968.

Holsboer F (1999) The rationale for the corticotropin-releasing hormone receptor (CRH-R) antagonists to treat depression and anxiety. J Psychiatr Res 33: 181-214.

Holsboer F (2000) The corticosteroid receptor hypothesis of depression. Neuropsycho- pharmacology 23: 477-501.

Holsboer F, Barden N (1996) Antidepressants and hypothalamic-pituitary-adrenocortical

regulation. Endocrine Rev 17: 187-205.

(8)

Holsboer F, Von Bardeleben U, Wiedemann K, Müller OA, Stalla GK (1987) Serial assessment of corticotropin-releasing hormone response after dexamethasone in depression:

implications for pathophysiology of DST nonsuppression. Biol Psychiat 22: 228-234.

Holsboer F, Lauer CJ, Schreiber W, Krieg J-C (1995) Altered hypothalamic-pituitary- adrenocortical regulation in healthy subjects at high familial risk for affective disorders.

Neuroendocrinology 62: 340-347.

Karssen AM, Meijer OC, van der Sandt ICJ, Lucassen PJ, de Lange EC, de Boer AG, de Kloet ER (2001) Multidrug resistance P-glycoprotein hampers the access of cortisol but not of corticosterone to mouse and human brain. Endocrinology 142: 2686-2694.

Keck ME, Holsboer F (2001) Hyperactivity of CRH neuronal circuits as a target for therapeutic interventions in affective disorders. Peptides 22: 835-844.

Keck ME, Welt T, Wigger A, Renner U, Engelmann M, Holsboer F, Landgraf R (2001) The anxiolytic effect of the CRH1 receptor antagonist R121919 depends on innate emotionality in rats. Eur J Neurosci 13: 373-380.

Lauer CJ, Schreiber W, Modell S, Holsboer F, Krieg JC (1998) The Munich vulnerability study on affective disorders: overview of the cross-sectional observations at index investigation.

J Psychiatric Res 32: 393-401.

Lucassen PJ, Müller MB, Holsboer F, Bauer J, Holtrop A, Wouda J, Hoogendijk WJG, de Kloet ER, Swaab DF (2001) Hippocampal apoptosis in major depression is a minor event and absent from subareas at risk for glucocorticoid overexposure. Am J Pathol 158: 453-468.

Malison RT, Anand A, Pelton GH, Kirwin P, Carpenter L, McDougle CJ, Heninger GR, Price LH (1999) Limited efficacy of ketoconazole in treatment-refractory major depression. J Clin Psychopharmacol 19: 466-470.

Mc Ewen BS (2000) The neurobiology of stress: from serendipity to clinical relevance. Brain Res 886: 172-189.

McQuade R, Young AH (2000) Future therapeutic targets in mood disorders: the glucocorticoid receptor. Br J Psychiat 177: 390-395.

Meijer OC, de Boer AG, van der Sandt ICJ, de Lange ECM, De Kloet ER (2000) The multidrug resistance 1A P-glycoprotein affects the penetration of glucocorticoids, except corticosterone, in the brain. Soc Neurosci Abstr 26: 419.

Mirow AL, Kristbjanarson H, Egeland JA, Shilling P, Helgason T, Gillin JC, Hirsch S, Kelsoe JR (1994) A linkage study of distal chromosome 5q and bipolar disorder. Biol Psychiat 36:

223-229.

Modell S, Yassouridis A, Huber J, Holsboer F (1997) Corticosteroid receptor function is decreased in depressed patients. Neuroendocrinology 65: 216-222.

Modell S, Lauer CJ, Schreiber W, Huber J, Krieg JC, Holsboer F (1998) Hormonal response pattern in the combined DEX-CRH test is stable over time in subjects at high familial risk for affective disorders. Neuropsychopharmacology 18: 253-262.

Morisette J, Villeenuve A, Lordeleau L, Rochette D, Laberge C, Gagné B, Laprise C, Bouchard G, Plante M, Gobeil L, Shink E, Weissenbach J, Barden N (1999) Genome-wide search for linkage of bipolar affective disorders in a very large pedigree derived from a homogenous population in Quebec points to a locus of major affect on chromosome 12q23-q24. Am J Med Genet 88: 567-587.

Moutsatsou P, Tsolakidou A, Trikkas G, Troungos C, Sekeris CE (2000) Glucocorticoid receptor alpha and beta isoforms are not mutated in bipolar affective disorder. Mol Psychiat 5: 196-202.

Müller MB, Keck ME (2002) Genetically engineered mice for studies of stress-related clinical conditions. J Psychiatr Res 36: 53-76.

Müller MB, Landgraf R, Sillaber I, Kresse AE, Keck ME, Zimmermann S, Holsboer F, Wurst W (2000) Selective activation of the hypothalamic vasopressinergic system in mice deficient for the corticotropin-releasing hormone receptor 1 is dependent on glucocorticoids.

Endocrinology 141: 4262-4269.

(9)

Müller MB, Lucassen PJ, Hoogendijk WGJ, Holsboer F, Swaab DF (2001) Neither major depression nor glucocorticoid treatment affects the cellular integrity of the human hippocampus. Eur J Neurosci 14: 1603-1612.

Müller MB, Holsboer F, Keck ME (2002) Genetic modification of corticosteroid receptor signalling: Novel insights into pathophysiology and treatment strategies of human affective disorders. Neuropeptides 36: 117-131.

Müller MB, Keck ME, Binder EB, Kresse AE, Hagemeyer TP, Landgraf R, Holsboer F , Uhr M (2003a) ABCB1- (MDR1)-type P-glycoproteins at the blood-brain barrier modulate the activity of the hypothalamic-pituitary-adrenocortical system: implications for affective disorder. Neurospychopharmacology 28: 1991-1999.

Müller MB, Zimmermann S, Sillaber I, Hagemeyer TP, Deussing JM, Timpl P, Kormann MSD, Droste S, Kühn R, Reul JMHM, Holsboer F, Wurst W (2003b) Limbic corticotropin- releasing hormone receptor 1 mediates anxiety-related behavior and hormonal adaptation to stress. Nature Neurosci 6: 1100-1107.

Murphy BE (1997) Antiglucocorticoid therapies in major depression: a review.

Psychoneuroendocrinology 22: 125-132.

Owens MJ, Nemeroff CB (1991) Physiology and pharmacology of corticotropin releasing factor. Pharmacol Rev 43: 425-473.

Purba JS, Hoogendijk WJG, Hofman MA, Swaab DF (1996) Increased number of vasopressin- and oxytocin-expressing neurons in the paraventricular nucleus of the hypothalamus in depression. Arch Gen Psychiat 53: 137-143.

Raadsheer FC, Hoogendijk WJG, Stam FC, Tilders FJH, Swaab DF (1994) Increased number of corticotropin-releasing hormone expressing neurons in the hypothalamic paraventricular nucleus of depressed patients. Neuroendocrinology 60: 436-444.

Ravaris CL, Sateia MJ, Beroza KW, Noordsy DL, Brinck-Johnsen T (1988) Effect of ketoconazole on a hypophysectomized, hypercortisolemic, psychotically depressed woman. Arch Gen Psychiat 45: 966-967.

Reul JMHM, de Kloet ER (1985) Two receptor systems for corticosterone in the rat brain:

microdistribution and differential occupation. Endocrinology 117: 2505-2512.

Rubin RT, Poland RE, Lesser IM, Winston RA, Blodgett ALN (1987) Neuroendocrine aspects of primary endogenous depression. Arch Gen Psychiat 44: 328-336.

Schulkin J, Gold PW, McEwen BS (1998) Induction of corticotropin-releasing hormone gene expression by glucocorticoids: implication for understanding the states of fear and anxiety and allostatic load. Psychoneuroendocrinology 23: 219-243.

Selye H (1946) The general adaptation syndrome and the diseases of adaptation. J Clin Endocrinol 6: 117-196.

Smith GW, Aubry J-M, Dellu F, Contarino A, Bilezikijan LM, Gold LH, Chen R, Marchuk Y, Hauser C, Bentley CA, Sawchenko PE, Koob GF, Vale W, Lee K-F (1998) Corticotropin releasing factor receptor 1-deficient mice display decreased anxiety, impaired stress response, and aberrant neuroendocrine development. Neuron 20: 1093-1102.

Syvanen AC (2001) Accessing genetic variation: genotyping single nucleotide polymorphisms.

Nature Rev Genet 2: 930-942.

Timpl P, Spanagel R, Sillaber I, Kresse A, Reul JMHM, Stalla GK, Blanquet V, Steckler T, Holsboer F, Wurst W (1998) Impaired stress response and reduced anxiety in mice lacking a functional corticotropin-releasing hormone receptor 1. Nature Genet 19: 162-166.

Uno H, Tarara R, Else JG, Suleman MA, Sapolsky RM (1989) Hippocampal damage associated with prolonged and fatal stress in primates. J Neurosci 9: 1705-1711.

Vale W, Spiess J, Rivier C, Rivier J (1981) Characterization of a 41-residue ovine hypothalamic peptide that stimulates secretion of corticotropin and b-endorphin. Science 213: 1394- 1397.

van Rossum EF, Koper JW, Huizenga NA, Uitterlinden AG, Janssen JA, O. BA, Grobbee DE, de

Jong FH, van Duyn CM, A. PH, Lamberts SW (2002) A polymorphism in the glucocorticoid

(10)

receptor gene, which decreases sensitivity to glucocorticoids in vivo, is associated with low insulin and cholesterol levels. Diabetes 51: 3128-3134.

Watts AG (1996) The impact of physiological stimuli on the expression of corticotropin- releasing hormone (CRH) and other neuropeptide genes. Front Neuroendocrinology 17:

281-326.

Wolkowitz OM, Reus VI, Chan T, Manfredi F, Raum W, Johnson R, Canick J (1999) Antiglucocorticoid treatment of depression: double-blind ketoconazole. Biol Psychiat 45: 1070-1074.

Zobel AW, Yassouridis A, Frieboes R-M, Holsboer F (1999) Prediction of medium-term outcome by cortisol response to the comined dexamethasone-CRH test in patients with remitted depression. Am J Psychiat 156: 949-951.

Zobel AW, Nickel T, Künzel HE, Ackl N, Sonntag A, Ising M, Holsboer F (2000) Effects of

the high-affinity corticotropin-releasing hormone receptor antagonist R121919 in major

depression: the first 20 patients treated. J Psychiatr Res 34: 171-181.

Riferimenti

Documenti correlati

These data indicate that bitopertin, by promoting heme restriction, ameliorates human β-thalassemic erythropoiesis, accelerates erythroid maturation, and reduces cell apoptosis..

Adults yielded the same trend in difficulty in making these rearrangements (Khemlani et al., 2013). When individuals rearrange cars, their moves are constrained by the goal –

An analysis of monthly average concentrations of total nitrogen and total phosphorus in the raw wastewater from the fruit and vegetable as well as dairy processing, reveals that

“In conclusion, our meta-analysis shows, that venous super-drainage, that is, performing a second venous anastomosis between the superficial venous system and a recipient vein,

In [6], which is the full version of [5], we propose a deterministic algorithm to compute a (finite) sound pre-fixed point of any effective realizer; however we have been able to

In this study, among 78 AF individuals of an age below 45 years without pre-existing pathologies and/or known risk factors, 13 (16.7%) had a type 1 BrS ECG pattern spontaneously (n =

Queste dif- ficoltà dei Monti nummari potevano avere diverse ragioni: innan- zitutto erano più soggetti all’insolvenza (mentre il grano si poteva recuperare facilmente,

A similar deletion was observed in case 11, whose array profile is illustrated in Papoulidis et al 19 ; B, Copy number profile of chromosome 1 from both cases as analyzed by