Skip to main content
Erschienen in: European Archives of Psychiatry and Clinical Neuroscience 8/2009

Open Access 01.12.2009 | Short Communication

Reduced hippocampal neurogenesis in the GR+/− genetic mouse model of depression

verfasst von: Golo Kronenberg, Imke Kirste, Dragos Inta, Sabine Chourbaji, Isabella Heuser, Matthias Endres, Peter Gass

Erschienen in: European Archives of Psychiatry and Clinical Neuroscience | Ausgabe 8/2009

Abstract

Glucocorticoid receptor (GR) heterozygous mice (GR+/ ) represent a valuable animal model for major depression. GR+/ mice show a depression-related phenotype characterized by increased learned helplessness on the behavioral level and neuroendocrine alterations with hypothalamo-pituitary-adrenal (HPA) axis overdrive characteristic of depression. Hippocampal brain-derived neurotrophic factor (BDNF) levels have also been shown to be reduced in GR+/ animals. Because adult hippocampal neurogenesis has been implicated in the pathophysiology of affective disorders, we studied here the effects of the GR+/ genotype on neurogenesis in vivo. In a 2 × 2 design, GR+/ mice and GR+/+ littermate controls were either subjected to 1 h of restraint stress or left undisturbed in their home cages after intraperitoneal injection of BrdU. Stress exposure and BrdU injections were performed once daily for 7 days and neurogenesis analyzed 4 weeks later. BrdU cell counts were significantly reduced as an effect of GR+/ genotype and as an effect of stress. Majority of the BrdU+ cells showed co-labeling with mature neuronal marker NeuN or astrocytic marker S100β with no further significant effect of either experimental condition or of genotype. In sum, this results in reduced neurogenesis in GR+/ mice which is further repressed by restraint stress. Our results, thus, reinforce the link between reduced neurogenesis, stress, neurotrophins, and behavioral symptoms of and susceptibility to depression.
Hinweise
G. Kronenberg and I. Kirste contributed equally.

Introduction

We have recently characterized a strain of glucocorticoid receptor (GR) heterozygous mice (GR+/ ) in which one GR allele has been ablated by a conventional knockout strategy, resulting in an approximately 50% reduction of GR protein levels in the brain [20]. These animals represent a valuable model for an endophenotype of severe major depression characterized by a disinhibited HPA-system due to altered GR function [11]. Similar to these severely depressed patients, GR+/ mice demonstrate a non-suppression of corticosterone plasma levels to dexamethasone and a pathological combined DEX/CRH test. Furthermore, GR+/ mice exhibit an altered neuroendocrine stress response with increased and prolonged corticosterone release after a stressor. Behaviorally, GR+/ mice show increased learned helplessness, a well-established analog of depression-like behavior in mice [4, 20].
As a molecular correlate—apart from reduced GR expression—for this ‘depressive’ phenotype we previously identified downregulation of BDNF at the protein level in hippocampus of GR+/ mice [20]. Compromised adult neurogenesis has also been implicated in the pathophysiology of hippocampal aspects of mood disorders [12]. Importantly, the ‘neurotrophin hypothesis’ of depression fits well with current concepts emphasizing the negative impact of stress on hippocampal structure [7]. Stress robustly impairs progenitor proliferation and neogenesis of granule neurons in the adult hippocampal dentate gyrus [5, 8, 9, 17]. Similarly, impaired BDNF signaling has recently been shown to result in impaired neurogenesis and refractory responses to antidepressants [15]. Deletion of the BDNF receptor trkB in adult progenitors also alters newborn neuron integration into hippocampal circuits and increases anxiety-like behaviors [1].
Here, we investigated net hippocampal neurogenesis in GR+/ mice either under control conditions or following repeated restraint stress. We speculated that neurogenesis may be compromised in this strain due to its depressive phenotype and concurrent BDNF downregulation.

Materials and methods

Animals and treatments

The generation of GR-heterozygous mice (GR+/ ) has been described in detail previously [20]. Briefly, the founders of GR+/ mice were developed by using homologous recombination in embryonic stem cells as described earlier [23]. GR+/ mice were generated by crossing heterozygous C57BL/6N males (backcrossed for >10 generations) with wild-type FVB/N females in order to obtain F1 hybrid mice with exactly the same background as previously used for behavioral and neurochemical characterization studies [20].
Male GR+/ mice and wild-type littermate controls were 4–6 months old at the beginning of experiments. Mice were housed individually in standard laboratory cages under a reversed 12 h:12 h light/dark cycle, with ad libitum access to food and water. All experiments were performed according to national and institutional guidelines and were approved by German animal welfare authorities. Thymidine analog BrdU (5-Bromo-2′-deoxyuridine; Sigma-Aldrich, Germany) was administered intraperitoneally at a concentration of 50 mg/kg body weight once daily. Restraint stress consisted of placing a mouse inside a plastic tube (inner diameter: 26 mm) for 1 h during the dark phase.
The experiment was structured in a 2 × 2 design (Fig. 1). Briefly, all animals received daily BrdU injections for a period of 7 days. Additionally, two groups were subjected to 1 h of restraint stress after BrdU injections. All animals were killed after an interval of 4 weeks. All experiments were performed according to the ‘Principles of laboratory animal care’ (NIH publication No. 86-23, revised 1985) and conformed to national and institutional guidelines.

Immunohistochemistry

Mice were deeply anesthetized with ketamine and transcardially perfused with 0.9% sodium chloride followed by 4% paraformaldehyde (PFA) in 0.1 M phosphate buffer. Brains were dissected from the skulls, postfixed in 4% PFA at 4 °C over night, and transferred into 30% sucrose for dehydration. Brains were cut on a sliding microtome (Leica, Bensheim, Germany) in the coronal plane in 40-μm-thick sections and cryoprotected. Sections were stained free floating with all antibodies diluted in Tris-buffered saline containing 3% donkey serum and 0.1% Triton X-100. For BrdU staining, DNA was denatured in 2 N HCl for 30 min at 37 °C. Sections were then rinsed in 0.1 M borate buffer and washed in Tris-buffered saline (TBS). Primary and secondary antibodies were diluted in TBS containing 0.1% Triton X-100 and 3% donkey serum (TBS+). Primary antibodies were applied in the following concentrations: anti-BrdU (rat, 1:500; Harlan Seralab, Indianapolis, IN), anti-NeuN (mouse, 1:100; Chemicon, Temecula, CA), anti-S100β (rabbit, 1:2500; Swant, Bellinzona, Switzerland). Immunohistochemistry followed the peroxidase method with biotinylated secondary antibody (Jackson ImmunoResearch Laboratories, West Grove, PA), ABC Elite reagent (Vector Laboratories, Burlingame, CA) and diaminobenzidine (DAB; Sigma) as chromogen. For immunofluorescence FITC-, RhodRedX- or Cy5-conjugated secondary antibodies were all used at a concentration of 1:250. Fluorescent sections were coverslipped in polyvinyl alcohol with diazabicyclooctane (DABCO) as an anti-fading agent.

Quantification and imaging

Cell counts were determined in one-in-six series of sections covering the entire hippocampus in its rostrocaudal extension as described previously [13, 14]. Briefly, cells located in the granule cell layer and adjacent subgranular zone, defined as a two-cell body-wide zone of the hilus along the base of the granule cell layer were counted [2]. Cells in the uppermost focal plane were excluded to avoid oversampling. Phenotypic analysis of BrdU-labeled cells was performed using a spectral confocal microscope (TCS SP2; Leica, Nussloch, Germany). Appropriate gain and black level settings were determined on control slices stained with secondary antibodies alone.

Statistical analyses

All numerical analyses were performed using Statview 5.0.1. for Macintosh. Two-way ANOVA was followed by Fisher’s post hoc test, where appropriate. All values are given as mean ± standard error of the mean (SEM). P-values of <0.05 were considered statistically significant.

Results

Net neurogenesis in the hippocampus was analyzed 4 weeks after a 7-day course of single daily intraperitoneal injections of BrdU. In this particular injection paradigm, BrdU counts reflect a combination of initial proliferation and subsequent survival of newly generated cells. The experimental set-up is illustrated in Fig. 1. Briefly, GR+/ mice and GR+/+ littermate controls were subjected either to the control condition (undisturbed in home cage) or to the stress condition (1 h of restraint stress directly after intraperitoneal injection of BrdU).
Two-way ANOVA revealed that BrdU cell counts were significantly reduced as an effect of stress and as an effect of GR+/ genotype (detailed results of the ANOVA statistics are presented in Figs. 2a and 3; analysis of 37 animals in total, n = 9–10 animals/group). BrdU-labeled cells were subjected to further phenotypic analysis. The majority of cells showed co-labeling with mature neuronal marker NeuN or astrocytic marker S100β (Figs.  2b and 4) with no further significant effect of either experimental condition or of genotype.

Discussion

Here we show that depression-prone GR+/ mice exhibit a significant net reduction of adult hippocampal neurogenesis. GR+/ mice represent a particularly valuable animal model of depression because they reflect HPA system dysregulation which is a hallmark of a subgroup of depressive disorders in humans [3]. Our observation is in line with the concept that neurogenesis has a role in the etiopathogenesis of mood disorders and contributes to the therapeutic actions of antidepressants [6, 16, 22]; but see also [10, 19, 24].
A number of current studies have provided novel insights into the link between depression-related behaviors and altered neurogenesis. In particular, recent work has shown that ‘learned safety’, as opposed to learned helplessness is associated with increased survival of new neurons and increased BDNF levels in hippocampus and that conversely, ablation of neurogenesis retards safety learning [18]. Several other recent studies also demonstrate a link between BDNF signaling, neurogenesis, and behavior [1, 15, 21]. In particular, ablation of BDNF receptor trkB in neural precursor cells results in impaired neurogenesis. When exposed to chronic antidepressant treatment or wheel-running, no increase in neurogenesis has been observed in these mice [15]. Furthermore, ablation of trkB also renders the animals behaviorally insensitive to antidepressive treatment in depression- and anxiety-like paradigms [15]. Lack of trkB in adult progenitors also results in disturbed organization of basic synaptic connections of newly generated neurons and impaired neurogenesis-dependent long-term potentiation, accompanied by compromised survival of newly generated cells and increased anxiety-like behaviors [1]. Reduced net hippocampal neurogenesis in GR+/ mice as reported here fits well with these reports. Importantly, we have previously demonstrated a significant reduction of BDNF protein concentrations in hippocampus of GR+/ mice [20]. However, it should be noted that the reduction in neurogenesis in GR+/ mice which emerged in this comparatively large experiment was only moderate. This may support the notion that in addition to reduced neurogenesis, other mechanisms may also contribute prominently to the depression-related behavioral phenotype of GR+/ mice [6].
In summary, we here demonstrate reduced net adult hippocampal neurogenesis in GR+/ mice, a novel genetic mouse model of affective disorders. Our study provides further correlative evidence for a link between hippocampal neurogenesis, glucocorticoids, BDNF, and depression.

Acknowledgments

This work was supported by the VolkswagenStiftung (Lichtenberg Program to M.E.), Bundesministerium für Bildung und Forschung (Center for Stroke Research Berlin; M.E.), Schilling Foundation (M.E.) and grants from the Deutsche Forschungsgemeinschaft (SFB636/B3 and GA 427/9-1 to P.G.).
Open Access This is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License ( https://​creativecommons.​org/​licenses/​by-nc/​2.​0 ), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Neuer Inhalt

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
1.
Zurück zum Zitat Bergami M, Rimondini R, Santi S, Blum R, Götz M, Canossa M (2008) Deletion of TrkB in adult progenitors alters newborn neuron integration into hippocampal circuits and increases anxiety-like behavior. Proc Natl Acad Sci USA 105(40):15570–15575CrossRefPubMed Bergami M, Rimondini R, Santi S, Blum R, Götz M, Canossa M (2008) Deletion of TrkB in adult progenitors alters newborn neuron integration into hippocampal circuits and increases anxiety-like behavior. Proc Natl Acad Sci USA 105(40):15570–15575CrossRefPubMed
2.
Zurück zum Zitat Brown J, Cooper-Kuhn CM, Kempermann G, Van Praag H, Winkler J, Gage FH, Kuhn HG (2003) Enriched environment and physical activity stimulate hippocampal but not olfactory bulb neurogenesis. Eur J Neurosci 17(10):2042–2046CrossRefPubMed Brown J, Cooper-Kuhn CM, Kempermann G, Van Praag H, Winkler J, Gage FH, Kuhn HG (2003) Enriched environment and physical activity stimulate hippocampal but not olfactory bulb neurogenesis. Eur J Neurosci 17(10):2042–2046CrossRefPubMed
3.
Zurück zum Zitat Chourbaji S, Gass P (2008) Glucocorticoid receptor transgenic mice as models for depression. Brain Res Rev 57(2):554–560CrossRefPubMed Chourbaji S, Gass P (2008) Glucocorticoid receptor transgenic mice as models for depression. Brain Res Rev 57(2):554–560CrossRefPubMed
4.
Zurück zum Zitat Chourbaji S, Zacher C, Sanchis-Segura C, Dormann C, Vollmayr B, Gass P (2005) Learned helplessness: validity and reliability of depressive-like states in mice. Brain Res Protoc 16:70–78CrossRef Chourbaji S, Zacher C, Sanchis-Segura C, Dormann C, Vollmayr B, Gass P (2005) Learned helplessness: validity and reliability of depressive-like states in mice. Brain Res Protoc 16:70–78CrossRef
5.
Zurück zum Zitat Czeh B, Welt T, Fischer AK, Erhardt A, Schmitt W, Muller MB, Toschi N, Fuchs E, Keck ME (2002) Chronic psychosocial stress and concomitant repetitive transcranial magnetic stimulation: effects on stress hormone levels and adult hippocampal neurogenesis. Biol Psychiatry 52:1057–1065CrossRefPubMed Czeh B, Welt T, Fischer AK, Erhardt A, Schmitt W, Muller MB, Toschi N, Fuchs E, Keck ME (2002) Chronic psychosocial stress and concomitant repetitive transcranial magnetic stimulation: effects on stress hormone levels and adult hippocampal neurogenesis. Biol Psychiatry 52:1057–1065CrossRefPubMed
6.
Zurück zum Zitat David DJ, Samuels BA, Rainer Q, Wang JW, Marsteller D, Mendez I, Drew M, Craig DA, Guiard BP, Guilloux JP, Artymyshyn RP, Gardier AM, Gerald C, Antonijevic IA, Leonardo ED, Hen R (2009) Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron 62(4):479–493CrossRefPubMed David DJ, Samuels BA, Rainer Q, Wang JW, Marsteller D, Mendez I, Drew M, Craig DA, Guiard BP, Guilloux JP, Artymyshyn RP, Gardier AM, Gerald C, Antonijevic IA, Leonardo ED, Hen R (2009) Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron 62(4):479–493CrossRefPubMed
7.
Zurück zum Zitat Gonul AS, Akdeniz F, Taneli F, Donat O, Eker C, Vahip S (2005) Effect of treatment on serum brain-derived neurotrophic factor levels in depressed patients. Eur Arch Psychiatry Clin Neurosci 255(6):381–386CrossRefPubMed Gonul AS, Akdeniz F, Taneli F, Donat O, Eker C, Vahip S (2005) Effect of treatment on serum brain-derived neurotrophic factor levels in depressed patients. Eur Arch Psychiatry Clin Neurosci 255(6):381–386CrossRefPubMed
8.
Zurück zum Zitat Gould E, McEwen BS, Tanapat P, Galea LA, Fuchs E (1997) Neurogenesis in the dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. J Neurosci 17:2492–2498PubMed Gould E, McEwen BS, Tanapat P, Galea LA, Fuchs E (1997) Neurogenesis in the dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. J Neurosci 17:2492–2498PubMed
9.
Zurück zum Zitat Gould E, Tanapat P, McEwen BS, Flugge G, Fuchs E (1998) Proliferation of granule cell precursors in the dentate gyrus of adult monkeys is diminished by stress. Proc Natl Acad Sci USA 95:3168–3171CrossRefPubMed Gould E, Tanapat P, McEwen BS, Flugge G, Fuchs E (1998) Proliferation of granule cell precursors in the dentate gyrus of adult monkeys is diminished by stress. Proc Natl Acad Sci USA 95:3168–3171CrossRefPubMed
10.
Zurück zum Zitat Holick KA, Lee DC, Hen R, Dulawa SC (2008) Behavioral effects of chronic fluoxetine in BALB/cJ mice do not require adult hippocampal neurogenesis or the serotonin 1A receptor. Neuropsychopharmacology 33(2):406–417CrossRefPubMed Holick KA, Lee DC, Hen R, Dulawa SC (2008) Behavioral effects of chronic fluoxetine in BALB/cJ mice do not require adult hippocampal neurogenesis or the serotonin 1A receptor. Neuropsychopharmacology 33(2):406–417CrossRefPubMed
11.
Zurück zum Zitat Holsboer F (2000) The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 23(5):477–501CrossRefPubMed Holsboer F (2000) The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 23(5):477–501CrossRefPubMed
12.
Zurück zum Zitat Kempermann G, Krebs J, Fabel K (2008) The contribution of failing adult hippocampal neurogenesis to psychiatric disorders. Curr Opin Psychiatry 21(3):290–295CrossRefPubMed Kempermann G, Krebs J, Fabel K (2008) The contribution of failing adult hippocampal neurogenesis to psychiatric disorders. Curr Opin Psychiatry 21(3):290–295CrossRefPubMed
13.
Zurück zum Zitat Kronenberg G, Lippoldt A, Kempermann G (2007) Two genetic rat models of arterial hypertension show different mechanisms by which adult hippocampal neurogenesis is increased. Dev Neurosci 29(1–2):124–133CrossRefPubMed Kronenberg G, Lippoldt A, Kempermann G (2007) Two genetic rat models of arterial hypertension show different mechanisms by which adult hippocampal neurogenesis is increased. Dev Neurosci 29(1–2):124–133CrossRefPubMed
14.
Zurück zum Zitat Kronenberg G, Reuter K, Steiner B, Brandt MD, Jessberger S, Yamaguchi M, Kempermann G (2003) Subpopulations of proliferating cells of the adult hippocampus respond differently to physiologic neurogenic stimuli. J Comp Neurol 467(4):455–463CrossRefPubMed Kronenberg G, Reuter K, Steiner B, Brandt MD, Jessberger S, Yamaguchi M, Kempermann G (2003) Subpopulations of proliferating cells of the adult hippocampus respond differently to physiologic neurogenic stimuli. J Comp Neurol 467(4):455–463CrossRefPubMed
15.
Zurück zum Zitat Li Y, Luikart BW, Birnbaum S, Chen J, Kwon CH, Kernie SG, Bassel-Duby R, Parada LF (2008) TrkB regulates hippocampal neurogenesis and governs sensitivity to antidepressive treatment. Neuron 59(3):399–412CrossRefPubMed Li Y, Luikart BW, Birnbaum S, Chen J, Kwon CH, Kernie SG, Bassel-Duby R, Parada LF (2008) TrkB regulates hippocampal neurogenesis and governs sensitivity to antidepressive treatment. Neuron 59(3):399–412CrossRefPubMed
16.
Zurück zum Zitat Malberg JE, Eisch AJ, Nestler EJ, Duman RS (2000) Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. J Neurosci 20(24):9104–9110PubMed Malberg JE, Eisch AJ, Nestler EJ, Duman RS (2000) Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. J Neurosci 20(24):9104–9110PubMed
17.
Zurück zum Zitat Pham K, Nacher J, Hof PR, McEwen BS (2003) Repeated restraint stress suppresses neurogenesis and induces biphasic PSA-NCAM expression in the adult rat dentate gyrus. Eur J Neurosci 17(4):879–886CrossRefPubMed Pham K, Nacher J, Hof PR, McEwen BS (2003) Repeated restraint stress suppresses neurogenesis and induces biphasic PSA-NCAM expression in the adult rat dentate gyrus. Eur J Neurosci 17(4):879–886CrossRefPubMed
18.
Zurück zum Zitat Pollak DD, Monje FJ, Zuckerman L, Denny CA, Drew MR, Kandel ER (2008) An animal model of a behavioral intervention for depression. Neuron 60(1):149–161CrossRefPubMed Pollak DD, Monje FJ, Zuckerman L, Denny CA, Drew MR, Kandel ER (2008) An animal model of a behavioral intervention for depression. Neuron 60(1):149–161CrossRefPubMed
19.
Zurück zum Zitat Reif A, Fritzen S, Finger M, Strobel A, Lauer M, Schmitt A, Lesch KP (2006) Neural stem cell proliferation is decreased in schizophrenia, but not in depression. Mol Psychiatry 11(5):514–522CrossRefPubMed Reif A, Fritzen S, Finger M, Strobel A, Lauer M, Schmitt A, Lesch KP (2006) Neural stem cell proliferation is decreased in schizophrenia, but not in depression. Mol Psychiatry 11(5):514–522CrossRefPubMed
20.
Zurück zum Zitat Ridder S, Chourbaji S, Hellweg R, Urani A, Zacher C, Schmid W, Zink M, Hörtnagl H, Flor H, Henn FA, Schütz G, Gass P (2005) Mice with genetically altered glucocorticoid receptor expression show altered sensitivity for stress-induced depressive reactions. J Neurosci 25(26):6243–6250CrossRefPubMed Ridder S, Chourbaji S, Hellweg R, Urani A, Zacher C, Schmid W, Zink M, Hörtnagl H, Flor H, Henn FA, Schütz G, Gass P (2005) Mice with genetically altered glucocorticoid receptor expression show altered sensitivity for stress-induced depressive reactions. J Neurosci 25(26):6243–6250CrossRefPubMed
21.
Zurück zum Zitat Rossi C, Angelucci A, Costantin L, Braschi C, Mazzantini M, Babbini F, Fabbri ME, Tessarollo L, Maffei L, Berardi N, Caleo M (2006) Brain-derived neurotrophic factor (BDNF) is required for the enhancement of hippocampal neurogenesis following environmental enrichment. Eur J Neurosci 24(7):1850–1856CrossRefPubMed Rossi C, Angelucci A, Costantin L, Braschi C, Mazzantini M, Babbini F, Fabbri ME, Tessarollo L, Maffei L, Berardi N, Caleo M (2006) Brain-derived neurotrophic factor (BDNF) is required for the enhancement of hippocampal neurogenesis following environmental enrichment. Eur J Neurosci 24(7):1850–1856CrossRefPubMed
22.
Zurück zum Zitat Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen R (2003) Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 301(5634):805–809CrossRefPubMed Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen R (2003) Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 301(5634):805–809CrossRefPubMed
23.
Zurück zum Zitat Tronche F, Kellendonk C, Kretz O, Gass P, Anlag K, Orban PC, Bock R, Klein R, Schutz G (1999) Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety. Nat Genet 23:99–103CrossRefPubMed Tronche F, Kellendonk C, Kretz O, Gass P, Anlag K, Orban PC, Bock R, Klein R, Schutz G (1999) Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety. Nat Genet 23:99–103CrossRefPubMed
24.
Zurück zum Zitat Vollmayr B, Mahlstedt MM, Henn FA (2007) Neurogenesis and depression: what animal models tell us about the link. Eur Arch Psychiatry Clin Neurosci 257(5):300–303CrossRefPubMed Vollmayr B, Mahlstedt MM, Henn FA (2007) Neurogenesis and depression: what animal models tell us about the link. Eur Arch Psychiatry Clin Neurosci 257(5):300–303CrossRefPubMed
Metadaten
Titel
Reduced hippocampal neurogenesis in the GR+/− genetic mouse model of depression
verfasst von
Golo Kronenberg
Imke Kirste
Dragos Inta
Sabine Chourbaji
Isabella Heuser
Matthias Endres
Peter Gass
Publikationsdatum
01.12.2009
Verlag
D. Steinkopff-Verlag
Erschienen in
European Archives of Psychiatry and Clinical Neuroscience / Ausgabe 8/2009
Print ISSN: 0940-1334
Elektronische ISSN: 1433-8491
DOI
https://doi.org/10.1007/s00406-009-0036-y

Weitere Artikel der Ausgabe 8/2009

European Archives of Psychiatry and Clinical Neuroscience 8/2009 Zur Ausgabe

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.

Weniger postpartale Depressionen nach Esketamin-Einmalgabe

Bislang gibt es kein Medikament zur Prävention von Wochenbettdepressionen. Das Injektionsanästhetikum Esketamin könnte womöglich diese Lücke füllen.

„Psychotherapie ist auch bei sehr alten Menschen hochwirksam!“

22.04.2024 DGIM 2024 Kongressbericht

Die Kombination aus Medikamenten und Psychotherapie gilt als effektivster Ansatz bei Depressionen. Das ist bei betagten Menschen nicht anders, trotz Besonderheiten.

Auf diese Krankheiten bei Geflüchteten sollten Sie vorbereitet sein

22.04.2024 DGIM 2024 Nachrichten

Um Menschen nach der Flucht aus einem Krisengebiet bestmöglich medizinisch betreuen zu können, ist es gut zu wissen, welche Erkrankungen im jeweiligen Herkunftsland häufig sind. Dabei hilft eine Internetseite der CDC (Centers for Disease Control and Prevention).

Update Psychiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.