Dear reader of ADxS.org, please excuse the disruption.

ADxS.org needs about $63500 in 2024. In 2023 we received donations of about $ 32200. Unfortunately, 99.8% of our readers do not donate. If everyone who reads this request makes a small contribution, our fundraising campaign for 2024 would be over after a few days. This donation request is displayed 23,000 times a week, but only 75 people donate. If you find ADxS.org useful, please take a minute and support ADxS.org with your donation. Thank you!

Since 01.06.2021 ADxS.org is supported by the non-profit ADxS e.V..

$8975 of $63500 - as of 2024-02-29
14%
Header Image
CRH

Sitemap

CRH

CRH, corticotropin releasing hormone, is also known as corticotropin releasing factor (CRF).

CRH is produced in the hypothalamus, the first stage of the HPA axis, and plays a key role in regulating the HPA axis and the human stress response.1

1. Formation and inhibition of CRH

1.1. Regions in which CRH is formed

  • In the paraventricular nucleus (PVN) of the hypothalamus (first stage of the HPA axis)
  • CRH is also produced in the limbic system and the cortex, among other places, and is likely to reinforce the effect of CRH from the hypothalamus on the pituitary gland.2
  • Furthermore, CRH is produced directly by peripheral nerves, where it is likely to have primarily immunological (inflammation-intensifying) effects.34

1.2. What increases CRH production

  • Stress
    • Stress results functionally in an increase in CRH5
      • In the central nucleus of the amygdala and
      • In the locus coeruleus
  • Hypoglycemia (low blood sugar)
  • Adrenalin6
  • Noradrenaline67
    • There are reciprocal (mutual) neuronal connections between CRH and noradrenergic cells in the locus coeruleus. CRH and noradrenaline thus stimulate each other, primarily via noradrenergic α1 receptors.83
      This enables the interaction between the HPA axis, the autonomic nervous system and the cardiovascular system to generate both short-term and long-term stress reactions.
  • Serotonin97
  • Acetylcholine7
  • Histamine10
  • Interleukin-1 (IL-1) of the monocytes11
  • Interleukin-6 (IL-6) of the monocytes11

1.3. What reduces CRH formation

  • Cortisol
    negative feedback loop - cortisol shuts down the HPA axis again
  • GABA9 and its agonists, such as benzodiapines10
  • CRH itself, via presynaptic CRH autoreceptors8

2. Effect of CRH

2.1. CRH receptors

CRH is 30 times more affine to CRH1 than to CRH2.12
Urocortin (UCN) has a high affinity for CRH1 and CRH212
UCN II and UCN III have a high affinity for CRH212

2.1.1. CRH1 receptor

CHR1 receptors mediate the CRH-controlled rapid response of the fight-flight system via the sympathetic nervous system and the HPA axis. CRH1 also activates the hypothalamic-pituitary-adrenal axis (HPA axis).13
In humans, CRH1 is thought to be more relevant in the brain than in the body.12

Highest density in

  • Cortex1415
    • CRH neurons in the anterior cingulate cortex (ACC) receive information from1
      • Cortex
      • Thalamus
      • Hippocampus
      • Amygdala
      • Several more midbrain and hindbrain nuclei
      • Basal forebrain cholinergic neurons
      • Serotonergic neurons of the raphe nuclei
  • Amygdala141
  • Cerebellum1415
  • Hippocampus14
  • Olfactory bulb14
  • Pituitary gland14
  • Thalamus16
  • Septum16
    • The CRH-1 receptor in the medial septum appears to mediate memory difficulties (more so in male than in female rats).17 CRH at the CRH-1 receptor in the medial septum mediated problems in retrieval but not in object recognition.
  • Substantia nigra16
  • Ventral tegmentum (VTA)16
  • Nucleus striae terminalis1
  • Paraventricular hypothalamic nucleus (PVN)1
  • Nucleus accumbens18

Mice without the CRH1 receptor have less anxiety and fewer cognitive disorders in stressful situations. Drugs that inhibit this receptor are currently being tested.1920

In a differentiating study on mice, it was found that CRH via CRH1 receptors

  • Triggers anxiety in glutamatergic systems of the forebrain and stimulates neurotransmission in the amygdala and hippocampus.
    Artificial selective removal of these CRH1 receptors reduced anxiety and inhibited neurotransmission in the amygdala and hippocampus.16
  • Stimulates the release of dopamine in dopaminergic cells in the ventral tegmentum and the substantia nigra, thereby reducing anxiety.
    Artificial selective removal of these CRH1 receptors reduced dopamine release in the PFC and increased anxiety.16

CRH release in the nucleus accumbens appears to play a key role in potentiating motivation for reward anticipation.21 Acute stress that increases CRH also increases dopamine, including in the nucleus accumbens, which triggers motivational expectancy. A CRH1 antagonist blocks this reinforcing effect of acute stress on reward motivation.22 High chronic stress abolishes - up to 90 days after the stressor has ended - the ability of CRH to increase dopamine in the nucleus accumbens and at the same time causes a switch from appetitive to aversive motivation,18 as is also observed in major depressive disorder MDD.23

It is possible that an imbalance between the glutamatergic and dopaminergic systems in relation to the effect of CRH contributes to the development of mental disorders.24

Intradermal CRH injection induces a marked increase in vascular permeability and mast cell degranulation mediated by CRH type 1 receptors.25

2.1.2. CRH2 receptor

The CRH2 receptor mediates the slower response of the stress system (adaptation and recovery). Urocortin II and urocortin III bind to CRH2.13 Urocortin is a neuropeptide related to CRH and binds much more strongly to CRH2 than CRH.14 It tends to have an anti-anxiety effect.13 The CRH2 receptor does not mediate symptoms of depression or anxiety, but stress scoping behavior.20 Unlike CRH1 receptors, cortisol does not inhibit the CRH effect at the CRH2 receptor.
In humans, CRH is thought to be more relevant in the body than in the brain.12

Highest density in

  • Hypothalamus14
  • Lateral septum1415
  • Hippocampus14
    • Ventral15
  • Olfactory bulb15
  • Bed nucleus of the stria terminalis15
  • Ventromedial hypothalamic nucleus15
  • Medial and posterior cortical nuclei of the amygdala15
  • Mesencephalic raphe nuclei15
  • And new localizations in the core of the solitary tract and area postrema.15
2.1.2.1. CRH2α receptor

In rats, the CRH2α receptor is not found in the brain.14

In humans, CRH2α is the predominant isoform. Found in both central and peripheral regions.
Mainly in subcortical brain regions:12

  • Hypothalamus
  • lateral septum
  • Olfactory bulb
2.1.2.2. CRH2β receptor

In rats, the CRH2β receptor is only found in the brain, but not the CRH2α receptor.14
Primarily in12

  • Heart
  • Skeletal muscles
2.1.2.3. CRH2-gamma receptor

Third isoform of the CRH2 receptor.12
Found in the amygdala in humans.

2.2. Neurophysiological effects of CRH

2.2.1. Activating effect of CRH

CRH activates or promotes

  • The (adeno-)pituitary gland (2nd stage of the HPA axis) by reducing the inhibitory potassium current26
  • The sympathetic nervous system (autonomic nervous system)27 by triggering the release of adrenaline in the adrenal medulla28
  • Dopamine synthesis29
  • Noradrenaline production in the locus coeruleus305
    • Noradrenaline controls the stress reactions of the brain (CNS).
    • Noradrenaline in turn activates the sympathetic nervous system and the HPA axis (positive feedback, reinforcement). This cascade is inhibited by GABA and glucocorticoids (cortisol).3132 Glucocorticoids inhibit CHR and noradrenaline production in the nucleus coeruleus.3
  • The release of somatostatin
    • Somatostatin in turn has an inhibitory effect on the HPA axis (negative feedback loop). This may be one reason for the frequent finding of elevated CRH levels with low cortisol levels.33
    • Somatostatin inhibits the secretion of
      • Growth hormone
      • TRH and
      • Thyrotropin
        and thus suppresses the reproductive, growth and thyroid functions, all of which are controlled by catecholamines.3

2.2.2. Inhibitory effect of CRH

CRH inhibits the development of:

  • GHRH3
  • Glycogen synthase kinase
    GSK-3β inactivates the protein glycogen synthase by phosphorylation and thereby contributes to the pathophysiology of Alzheimer’s disease. The use of CRH in the treatment of Alzheimer’s disease is discussed.34

2.3. Behavioral effect of CRH

2.3.1. Behavioral triggering by CRH

CRH (injected directly) triggers immediate stress reactions.35 CRH antagonists block these reactions.27

  • Attention
    • Sustained attention is impaired by CRH.36
  • Acoustic perception
  • Memory problems
    • CRH in the medial septum (which projects into the hippocampus) caused memory problems in the hippocampus in both male and female rats. Males were more sensitive to CRH. Females had higher levels of CRH-binding protein, which reduced CRH levels. CRH1 receptors were equally strong in both sexes. CRH1 antagonists prevented the memory problems caused by CRH.37
  • Alertness
  • Locomotor activity (urge to move)538
  • Take care in unknown environments, in open spaces, in the elevated plus maze and in the event of conflicts
  • Exploration intensity in a known environment38
  • Reduced exploration activity in a new environment12
  • Freezing behavior in an unfamiliar environment38
  • Social withdrawal behavior (promotes social phobia)
  • Fear12
    • A differentiating study in mice found that CRH via CRHR1 receptors
      • Triggers anxiety in glutamatergic systems of the forebrain and stimulates neurotransmission in the amygdala and hippocampus.
        Artificial selective removal of these CRH1 receptors reduced anxiety and inhibited neurotransmission in the amygdala and hippocampus.16
      • Stimulates the release of dopamine in dopaminergic cells in the ventral tegmentum and the substantia nigra, thereby reducing anxiety.
        Artificial selective removal of these CRHR1 receptors reduced dopamine release in the PFC and increased anxiety.16
    • CRH mediates escape behavior and fear conditioning in the presence of unavoidable pain by activating the release of serotonin in the (caudal dorsal) raphe nuclei (DRN). CRH, which is applied directly to the caudal DRN, mediates these reactions even without unavoidable inflictions of pain.39 CRH in the rostral DRN did not trigger this.
  • Startle reaction
  • Fear conditioning39
    • Increased aversion to a stressor due to the fear experienced as a result of it40
  • Despair
  • Negative mood41
  • Increased thermogenesis via the catecholaminergic system3
  • Increase in energy turnover and fat burning34
  • Increased heart rate and blood pressure12
  • Stimulation/inhibition of gastrointestinal functions12
  • Reduced sleep12
  • Reduced food intake12

2.3.2. Behavioral inhibition due to CRH

CRH reduced:

  • Appetite via the catecholaminergic system3
  • Libido
  • Slow-wave sleep / deep sleep
  • CRH (when applied directly to the dorsal mPFC, where a particularly large number of CRH receptors are located, and also when applied globally in the brain) reduces the performance of the PFC (especially working memory) in a dose-dependent manner. CRH antagonists neutralize this effect.4219
    Working memory is particularly severely impaired in ADHD.

2.3.3. CRH and mental disorders

CRH is elevated in1

  • Alzheimer’s disease
  • Depression
  • Anxiety disorders

2.4. Immunological effect of CRH

  • CRH secreted directly by the peripheral nerves stimulates local inflammation (immune CRH)34
  • CRH in the brain inhibits the inflammatory reaction via glucocorticoids (cortisol) and catecholamines.3
    This presumably means that CRH triggers a cortisol reaction. Cortisol is known to inhibit inflammation.

2.5. Neurotoxic effects of CRH in chronic stress

Prolonged high CRH levels lead to a reduced binding capacity of the CRH receptors in the PFC (downregulation).43 As the PFC is involved in the inhibition of the HPA axis (stress axis), a long-lasting high cortisol level leads to an impairment of the inhibition of the HPA axis.44

The effect of CRH can be enhanced by vasopressin on the corticotrophic hypohyseal cells, especially during prolonged stress.45
CRH release is regulated by higher-level instances, including the hippocampus.45

3. CRH agonists and antagonists as medication

3.1. Areas of application of CRH agonists

In the case of a hypoactive HPA axis, which can usually be identified by a flattened cortisostress response, fatigue, depressive symptomatology, hyperalgesia and increased immune / inflammatory reactions, CRH agonists could possibly be helpful. Areas of application could be, for example

  • Atypical depression3
  • Bipolar disorder / manic-depressive disorder
    In our opinion, this results from the fact that bipolar depression, like atypical depression, is characterized by a flattened endocrine stress response.
  • Postpartum depression3
  • Fibromyalgia3
  • Chronic exhaustion syndrome (fatigue)3
  • ADHD-HI (with hyperactivity)
    In our opinion, this results from the fact that ADHD-HI is also characterized by a flattened endocrine stress response.

3.2. Areas of application of CRH antagonists

In the case of a hyperactive HPA axis, which can usually be identified by an excessive cortisol stress response, CRH antagonists could possibly be helpful.

CRH antagonists are for example:

  • Alpha-helical CRH (ah-CRH)12
  • D-Phe CRH (d-Phe CRH)12
  • Antalarmin (CP-156,181); similar to CP-154,526)
    • Selective CRH-1 antagonist12
    • 20 mg / kg in primates exposed to social stress caused46
      • Reduced anxiety / fear behavior
        • Body tremors
        • Grimace
        • Teeth grinding
        • Urinating
        • Bowel movement
      • Reinforced behavior that is suppressed under stress
        • Explorative behavior
        • Sexual behavior
      • Antalarmin (also CP-154,526) prevent ACTH increase on CRH12
        • Basal ACTH level
          • Unchanged after 1 week
          • Reduced after 11 days as well as after 8 weeks (corticosterone also reduced)
            • Reduced responsiveness of the adrenal cortex to ACTH
            • No signs of adrenal insufficiency
          • ACTH and corticosterone response to immobilization stress unchanged
            • Response to acute stress remained intact
      • Antalarmin inhibited ACTH response to conditioned fear and social stress, but not to inescapable pain shocks12
  • CP-154,526
    • Antalarmin analog
    • Selective CRH-1 antagonist12
    • Low oral bioavailability
    • High hepatic clearance
    • Drug development was stopped preclinically
    • CP-154,526 inhibited ACTH response to immobilization stress, but less to other stressors such as cold stress
    • CP-154,526 applied to the locus coeruleus inhibited noradrenaline response to handling stress, but left noradrenaline, dopamine and serotonin levels in the PFC unchanged
  • Astressin12

    • CRH1 and CRH2 antagonist
  • Antisauvagin-30 (AS-30)12

    • Over 300-fold selectivity for CRH2 compared to CRH1
  • R121919 (NBI30775)47

    • Selective CRH1 antagonist (Ki 2 - 5 nM)
    • CRH2 binding over 1000 times weaker
  • SSR125543A

    • CRH1 antagonist
  • Alprazolam

    • Significantly inhibited serotonin-induced CRH release in a dose-dependent manner48
    • Probably due to inhibition of CRH secretion
  • Diazepam

    • Significantly inhibited serotonin-induced CRH release in a dose-dependent manner, but 40 times weaker than alprazolam48

Areas of application could be, for example

  • Melancholic depression3
  • Psychotic depression
  • Chronic anxiety3
  • ADHD-I
    In our opinion, this results from the fact that ADHD-I is also characterized by an excessive endocrine stress response.
  • SCT
    In our opinion, this results from the fact that SCT is most likely also characterized by a flattened endocrine stress response.

4. Measurement of CRH

CRH in the brain can only be measured in the cerebrospinal fluid. This is not practical for standard diagnostics or treatment.
CRH can also be measured in blood plasma.

5. CRH and ADHD

One study found that CRH1 antagonists were able to improve working memory in rats stressed by intensive handling in a dose-dependent manner. The improvement corresponded to the degree of improvement with ADHD medication.49 This applied to the non-selective CRH antagonist D-Phe-CRF as well as the selective CRH1 antagonist NBI 35965.


  1. Zhang, Lv, Yuan, Fan, Li, Sun, Hu (2019): Whole-Brain Mapping of Monosynaptic Afferent Inputs to Cortical CRH Neurons. Front Neurosci. 2019 Jun 4;13:565. doi: 10.3389/fnins.2019.00565. eCollection 2019.

  2. Rensing, Koch, Rippe, Rippe (2006): Mensch im Stress; Psyche, Körper Moleküle; Elsevier (jetzt Springer), Seite 95

  3. Tsigos, Chrousos (2002): Hypothalamic–pituitary–adrenal axis, neuroendocrine factors and stress; Journal of Psychosomatic Research, Volume 53, Issue 4, 2002, Pages 865-871, ISSN 0022-3999, https://doi.org/10.1016/S0022-3999(02)00429-4.

  4. Karalis, Sano, Redwine, Listwak, Wilder, Chrousos (1991): Autocrine or paracrine inflammatory actions of corticotropin-releasing hormone in vivo. Science 18 Oct 1991: Vol. 254, Issue 5030, pp. 421-423; DOI: 10.1126/science.1925600

  5. Egle, Joraschky, Lampe, Seiffge-Krenke, Cierpka (2016): Sexueller Missbrauch, Misshandlung, Vernachlässigung – Erkennung, Therapie und Prävention der Folgen früher Stresserfahrungen; 4. Aufl., Schattauer, S. 46

  6. Plotsky, Cunningham, Widmaier (1989): Catecholaminergic modulation of corticotropin-releasing factor and adrenocorticotropin secretion. Endocr Rev. 1989 Nov;10(4):437-58.

  7. Birbaumer, Schmidt (2010): Biologische Psychologie. Berlin/Heidelberg: Springer

  8. Chida, Hamer (2008): Chronic Psychosocial Factors and Acute Physiological Responses to Laboratory-Induced Stress in Healthy Populations: A Quantitative Review of 30 Years of Investigations; Psychological Bulletin 2008, Vol. 134, No. 6, 829–885 0033-2909/08/$12.00 DOI: 10.1037/a0013342

  9. Assenmacher I, Szafarczyk, Alonso, Ixart, Barbanel (1987): Physiology of neural pathways affecting CRH secretion. In: Ganong, Dallman, Roberts (eds): The hypothalamic-pitutary-adrenal axis revisited. Ann N Y Acad Sci, 1987, 512: 149-161

  10. Calogero (1995): Neurotransmitter regulation of the hypothalamic corticotropinreleasing hormone neuron. Ann N Y Acad Sci, 771, 31-40

  11. Gutscher (2002): Der Glucocorticoidrezeptor des Schweins: Herstellung und Charakterisierung eines polyklonalen Antiserums. sowie Studien zur Verteilung des GCR im Testinaltrakt von Ebern und Kastraten, Dissertation, Seite 16 mwNw

  12. Seymour, Schmidt, Schulz (2003): The pharmacology of CP-154,526, a non-peptide antagonist of the CRH1 receptor: a review. CNS Drug Rev. 2003 Spring;9(1):57-96. doi: 10.1111/j.1527-3458.2003.tb00244.x. PMID: 12595912; PMCID: PMC6741649.

  13. de Kloet, Joëls, Holsboer (2005): Stress and the brain: from adaptation to disease. Nat Rev Neurosci. 2005 Jun;6(6):463-75.

  14. Wagner, Born: Psychoendokrine Aspekte neurophysiologischer Funktionen. In: Lautenbacher, Gauggel (2013): Neuropsychologie psychischer Störungen, Springer, Seite 125

  15. Van Pett, Viau, Bittencourt, Chan, Li, Arias, Prins, Perrin, Vale, Sawchenko (2000): Distribution of mRNAs encoding CRF receptors in brain and pituitary of rat and mouse. J Comp Neurol. 2000 Dec 11;428(2):191-212. doi: 10.1002/1096-9861(20001211)428:2<191::aid-cne1>3.0.co;2-u. PMID: 11064361.

  16. Refojo, Schweizer, Kuehne, Ehrenberg, Thoeringer, Vogl, Dedic, Schumacher, von Wolff, Avrabos, Touma, Engblom, Schütz, Nave, Eder, Wotjak, Sillaber, Holsboer, Wurst, Deussing (2011): Glutamatergic and Dopaminergic Neurons Mediate Anxiogenic and Anxiolytic Effects of CRHR1; Science Express, DOI:10.1126/science.1202107

  17. Wiersielis, Ceretti, Hall, Famularo, Salvatore, Ellis, Jang, Wimmer, Bangasser (2019): Sex differences in corticotropin releasing factor regulation of medial septum-mediated memory formation. Neurobiol Stress. 2019 Feb 20;10:100150. doi: 10.1016/j.ynstr.2019.100150. PMID: 30937355; PMCID: PMC6430617.

  18. Lemos, Wanat, Smith, Reyes, Hollon, Van Bockstaele, Chavkin, Phillips (2012): Severe stress switches CRF action in the nucleus accumbens from appetitive to aversive. Nature. 2012 Oct 18;490(7420):402-6. doi: 10.1038/nature11436. PMID: 22992525; PMCID: PMC3475726.

  19. http://www.depression-therapie-forschung.de/hormone.html

  20. Heim, Miller: Depression, in: Ehlert, von Känel (2011): Psychoendokrinologie und Psychoimmunologie, Seiten 365-382

  21. Peciña, Schulkin, Berridge (2006): Nucleus accumbens corticotropin-releasing factor increases cue-triggered motivation for sucrose reward: paradoxical positive incentive effects in stress? BMC Biol. 2006 Apr 13;4:8. doi: 10.1186/1741-7007-4-8. PMID: 16613600; PMCID: PMC1459217.

  22. Liu (2015): Enhanced motivation for food reward induced by stress and attenuation by corticotrophin-releasing factor receptor antagonism in rats: implications for overeating and obesity. Psychopharmacology (Berl). 2015 Jun;232(12):2049-60. doi: 10.1007/s00213-014-3838-1. PMID: 25510859; PMCID: PMC4433618.

  23. Ironside, Kumar, Kang, Pizzagalli (2018): Brain mechanisms mediating effects of stress on reward sensitivity. Curr Opin Behav Sci. 2018 Aug;22:106-113. doi: 10.1016/j.cobeha.2018.01.016. PMID: 30349872; PMCID: PMC6195323.

  24. Refojo, Deussing (2012): Das Corticotropin-Releasing-Hormon-System und die Angst; Wissenschaft Molekulare Medizin; February 2012, Volume 18, Issue 1, pp 15–18

  25. Theoharides, Singh, Boucher, Pang, Letourneau, Webster, Chrousos (1998): Corticotropin-Releasing Hormone Induces Skin Mast Cell Degranulation and Increased Vascular Permeability, A Possible Explanation for Its Proinflammatory Effects, Endocrinology, Volume 139, Issue 1, 1 January 1998, Pages 403–413, https://doi.org/10.1210/endo.139.1.5660

  26. Aldenhoff, Erregungsungleichgewicht als mögliche Ursache seelischer Erkrankungen (1990) in Beckmann, Osterheider: Neurotransmitter und psychische Erkrankungen, Springer, Seite 183

  27. Egle, Joraschky, Lampe, Seiffge-Krenke, Cierpka (2016): Sexueller Missbrauch, Misshandlung, Vernachlässigung – Erkennung, Therapie und Prävention der Folgen früher Stresserfahrungen; 4. Aufl., Schattauer, S. 45

  28. Bruhn, Engeland,Anthony, Gann, Jackson (1987): Corticotropin‐releasing Factor in the Adrenal Medulla. Annals of the New York Academy of Sciences, 512: 115-128. doi:10.1111/j.1749-6632.1987.tb24954.x

  29. Payer, Williams, Mansouri, Stevanovski, Nakajima, Le Foll, Kish, Houle, Mizrahi, George, George, Boileau (2017): Corticotropin-releasing hormone and dopamine release in healthy individuals. Psychoneuroendocrinology. 2017 Feb;76:192-196. doi: 10.1016/j.psyneuen.2016.11.034. PMID: 27951520.

  30. Rensing, Koch, Rippe, Rippe (2006): Mensch im Stress; Psyche, Körper Moleküle; Elsevier (jetzt Springer), Seite 96

  31. Egle, Joraschky, Lampe, Seiffge-Krenke, Cierpka (2016): Sexueller Missbrauch, Misshandlung, Vernachlässigung – Erkennung, Therapie und Prävention der Folgen früher Stresserfahrungen; 4. Aufl., Schattauer, S. 47

  32. Chida, Hamer (2008): Chronic Psychosocial Factors and Acute Physiological Responses to Laboratory-Induced Stress in Healthy Populations: A Quantitative Review of 30 Years of Investigations, Psychological Bulletin 2008, Vol. 134, No. 6, 829–885 0033-2909/08/$12.00 DOI: 10.1037/a0013342

  33. Newport, Stowe, Nemeroff (2002): Parental Depression: Animal Models of an Adverse Life Event; American Journal of Psychiatry 2002 159:8, 1265-1283

  34. Strohner (2011): Auswirkungen des Polymorphismus rs3176921 im CRH-Gen auf kognitive Phänotypen; Dissertation

  35. Rensing, Koch, Rippe, Rippe (2006): Mensch im Stress; Psyche, Körper Moleküle; Elsevier (jetzt Springer), Seite 96, Seite 151

  36. Hupalo, Spencer, Berridge (2021): Prefrontal corticotropin-releasing factor neurons impair sustained attention via distal transmitter release. Eur J Neurosci. 2021 May 5. doi: 10.1111/ejn.15260. PMID: 33949025.

  37. Wiersielis, Ceretti, Hall, Famularo, Salvatore, Ellis, Jang, Wimmer, Bangasser (2019): Sex differences in corticotropin releasing factor regulation of medial septum-mediated memory formation. Neurobiol Stress. 2019 Feb 20;10:100150. doi: 10.1016/j.ynstr.2019.100150.

  38. Arborelius, Owens, Plotsky, Nemeroff (1999): The role of corticotropin-releasing factor in depression and anxiety disorders. J Endocrinol, 160(1), 1-12.

  39. Hammack, Richey, Schmid, LoPresti, Watkins, Maier (2002): The Role of Corticotropin-Releasing Hormone in the Dorsal Raphe Nucleus in Mediating the Behavioral Consequences of Uncontrollable Stress; Journal of Neuroscience 1 February 2002, 22 (3) 1020-1026; DOI: https://doi.org/10.1523/JNEUROSCI.22-03-01020.2002

  40. Holsboer, Ising (2010): Stress Hormone Regulation: Biological Role and Translation into Therapy. Annual Review of Psychology 2010 61:1, 81-109

  41. Lautenbacher, Gauggel (2013): Neuropsychologie psychischer Störungen, Springer, Seite 141

  42. Hupalo, Berridge (2016): Working Memory Impairing Actions of Corticotropin-Releasing Factor (CRF) Neurotransmission in the Prefrontal Cortex. Neuropsychopharmacology. 2016 Oct;41(11):2733-40. doi: 10.1038/npp.2016.85.

  43. Arborelius, Owens, Plotsky, Nemeroff (1999): The role of corticotropin-releasing factor in depression and anxiety disorders. J Endocrinol 1999; 160: 1–12, Seite 5

  44. Egle, Joraschky, Lampe, Seiffge-Krenke, Cierpka (2016): Sexueller Missbrauch, Misshandlung, Vernachlässigung – Erkennung, Therapie und Prävention der Folgen früher Stresserfahrungen; 4. Aufl., Schattauer, S. 44

  45. Trapp, Holzboer (2013): Molekulare Mechanismen der Glucocorticoidtherapie; in: Ganten, Ruckpaul (2013): Erkrankungen des Zentralnervensystems, Springer, Seite 104

  46. Habib, Weld, Rice, Pushkas, Champoux, Listwak, Webster, Atkinson, Schulkin, Contoreggi, Chrousos, McCann, Suomi, Higley, Gold (2000): Oral administration of a corticotropin-releasing hormone receptor antagonist significantly attenuates behavioral, neuroendocrine, and autonomic responses to stress in primates. Proc Natl Acad Sci U S A. 2000 May 23;97(11):6079-84. doi: 10.1073/pnas.97.11.6079. PMID: 10823952; PMCID: PMC18561.

  47. Skelton, Oren, Gutman, Easterling, Holtzman, Nemeroff, Owens (2007): The CRF1 receptor antagonist, R121919, attenuates the severity of precipitated morphine withdrawal. Eur J Pharmacol. 2007 Sep 24;571(1):17-24. doi: 10.1016/j.ejphar.2007.05.041. PMID: 17610870.

  48. Kalogeras, Calogero, Kuribayiashi, Khan, Gallucci, Kling, Chrousos, Gold (1990): In vitro and in vivo effects of the triazolobenzodiazepine alprazolam on hypothalamic-pituitary-adrenal function: pharmacological and clinical implications. J Clin Endocrinol Metab. 1990 May;70(5):1462-71. doi: 10.1210/jcem-70-5-1462. PMID: 2159487.

  49. Hupalo, Berridge (2016): Working Memory Impairing Actions of Corticotropin-Releasing Factor (CRF) Neurotransmission in the Prefrontal Cortex. Neuropsychopharmacology. 2016 Oct;41(11):2733-40. doi: 10.1038/npp.2016.85. Epub 2016 Jun 8. PMID: 27272767; PMCID: PMC5026742.

Diese Seite wurde am 27.02.2024 zuletzt aktualisiert.