Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jul 18.
Published in final edited form as: Psychoneuroendocrinology. 2009 Dec;34(Suppl 1):S84–S90. doi: 10.1016/j.psyneuen.2009.06.019

Steroid Hormone Fluctuations and GABAAR Plasticity

Jamie Maguire 1, Istvan Mody 1
PMCID: PMC3399241  NIHMSID: NIHMS134559  PMID: 19632051

Abstract

Conditions of changing steroid hormone levels are a particularly vulnerable time or the manifestation of neurological disorders, including catamenial epilepsy, premenstrual syndrome (PMS), and postpartum depression. The pathophysiology of these disorders may be related to changes in neurosteroid levels, which can dramatically impact neuronal excitability. Robust changes in neurosteroid levels, such as those that occur following stress, over the ovarian cycle, and throughout pregnancy, profoundly alter GABAA receptor (GABAAR) structure and function and underlie the associated changes in neuronal excitability. A moderate and brief exposure to elevated neurosteroids, such as those that occur over the ovarian cycle and following an acute stressful episode, result in a decrease in GABAAR γ2 subunit expression and an increase in GABAAR δ subunit expression. These changes are accompanied by a decrease in seizure susceptibility and decreased anxiety-like behavior in mice, demonstrating altered neuronal excitability associated with changes in the receptor composition. More robust changes in steroid hormone levels, such as those that occur throughout pregnancy, result in a decrease in both GABAAR γ2 and δ subunit expression and are associated with an increase in neuronal excitability evident from the shift in the input-output relationship. Alterations in GABAAR subunit composition may represent a homeostatic mechanism to maintain an ideal level of inhibition in the face of fluctuating neurosteroid levels. Neurosteroids potentiate the effects of GABA on GABAARs, particularly those containing the δ subunit, and reorganization of these receptors may be necessary to prevent sedation and/or anaesthesia in the face of high levels of neurosteroids or to prevent hyperexcitability in the absence of these compounds. Alterations in GABAARs under conditions of altered steroid hormone levels result in measurable changes in neuronal excitability and dysregulation of GABAARs may play a role in steroid hormone-associated neurological disorders.

Neurosteroids in the Central Nervous System (CNS)

Neurosteroids are neuroactive metabolites of steroid hormones, which can be synthesized in the peripheral and central nervous system, in both neurons and glial cells (Belelli and Lambert, 2005;Herd et al., 2007). Neurosteroids are synthesized de novo from cholesterol or converted from steroid hormone precursors (Stoffel-Wagner, 2001). Thus, fluctuations in steroid hormone levels result in local fluctuations in neurosteroid levels (Stoffel-Wagner, 2001). Altered levels of neurosteroids in the CNS are associated with numerous physiological (e.g., ovarian cycle, pregnancy) and pathological (e.g., stress) conditions. Some of these conditions are known to be associated with debilitating psychiatric and neurological disorders including premenstrual dysphoric disorder (PMDD), premenstrual syndrome (PMS), catamenial epilepsy, menstrual migraine, postpartum depression, and anxiety (for review see (Backstrom et al., 2003)).

Steroid hormone-linked disorders in women share patterns of symptom manifestation, consisting of a worsening during the luteal phase, occurring just prior to or during menses, and/or at the time of ovulation (Backstrom et al., 2003;Smith et al., 2003;Sundstrom et al., 1999). Symptom manifestation in menstrual cycle-linked psychiatric and neurological disorders has been attributed to changes in steroid hormone levels. However, there are no consistent changes in hormone levels associated with steroid hormone-linked disorders (for review see (Backstrom et al., 2003)). Rather, it has been suggested that women with PMS/PMDD differ in brain sensitivity to neurosteroids (Backstrom et al., 2003). Administration and withdrawal of exogenous steroid hormones, designed to mimic the hormonal changes during pregnancy, result in symptoms of depression only in women with a history of postpartum depression (Bloch et al., 2000), suggesting that the withdrawal of reproductive hormones alone is not sufficient to induce symptoms of depression, but rather some women must be predisposed to postpartum depression. Consistent with this hypothesis, women with a history of postpartum depression exhibit differences in their sensitivities to steroid hormones (Wisner et al., 2002), suggesting that the site of neurosteroid action, namely the GABAA receptors (GABAARs), might be altered in affected women.

Neurosteroids and GABAA Receptors

The GABAergic system has been implicated in the pathophysiology of menstrual cycle-linked disorders (Backstrom et al., 2003;Smith et al., 2003;Sundstrom et al., 1999) as well as anxiety disorders (Bremner et al., 2000;Goddard et al., 2001;Malizia et al., 1998;Tiihonen et al., 1997). Benzodiazepines and the neuroactive steroids pregnanolone and allopregnanolone exert anxiolytic effects in patients with panic disorder (Herd et al., 2007;Mitchell et al., 2008;Smith et al., 2007;Strohle et al., 2002). In addition, withdrawal of positive allosteric modulators of GABAARs, such as benzodiazepines, mimic the symptoms of ovarian cycle-linked neurological disorders, resulting in symptoms such as irritability, tension, increased anxiety, panic attacks, headaches, and even depression and seizures in women (Petursson, 1994). Consistent with the role of GABAARs in menstrual cycle-linked disorders, PMS/PMDD patients are less sensitive to neurosteroids and benzodiazepines (Sundstrom et al., 1997;Sundstrom et al., 1998).

GABAARs are heteropentameric receptors composed of a combination of subunits, such as α1-6, β1-4, γ1-3,δ, ε, θ, and ρ1-2 (Olsen and Sieghart, 2008;Whiting et al., 1999), which dictate the anatomical distribution (Pirker et al., 2000), physiological properties, and pharmacology of the receptor (Hevers and Luddens, 1998;Mody and Pearce, 2004). For example, γ2 containing GABAARs are widely distributed throughout the brain (Pirker et al., 2000;Wisden et al., 1992). These receptors are localized synaptically and mediate the “synaptic” or “phasic” form of GABAergic inhibition (Farrant and Nusser, 2005). In contrast, the δ subunit containing GABAARs are prevalent in the cerebellum, dentate gyrus, cortex, thalamus, and striatum (Peng et al., 2002;Peng et al., 2004;Pirker et al., 2000;Wisden et al., 1992). GABAAR δ subunit-containing receptors are localized extrasynaptically (Nusser et al., 1998) or perisynaptically (Wei et al., 2003) and mediate the “tonic” form of GABAergic inhibition (Farrant and Nusser, 2005). We now know δ subunit-containing GABAARs are more sensitive to neurosteroids in heterologous systems (Belelli et al., 2002;Belelli and Lambert, 2005;Brown et al., 2002;Mihalek et al., 1999;Spigelman et al., 2003;Wohlfarth et al., 2002) and these receptors are the major site of neurosteroid potentiation of the tonic current in dentate gyrus granule cells (Stell et al., 2003) (for review see (Walker and Semyanov, 2008)). However, the conserved site for neurosteroid binding has recently been discovered between the α and β interface of the GABAAR (Hosie et al., 2006), in conflict with the pharmacological data demonstrating the enhanced neurosteroid potentiation of δ subunit-containing receptors. This discrepancy may be due to the fact that the δ subunit may act to alter the efficacy of neurosteroid potentiation rather than influence neurosteroid binding (Hosie et al., 2009).

GABAA Receptor Regulation by Steroid Hormones

While steroid hormone metabolites, derived from ovarian steroid hormones (progesterone) or stress steroid hormones (corticosterone), potentiate the effects of GABA on GABAARs, particularly those containing δ subunits, there are also effects of steroid hormones and neurosteroids on GABAAR subunit expression in rodents. the remainder of this review will focus on the steroid hormone regulation of GABAAR structure and function in response to physiological conditions, such as pregnancy, stress, and the ovarian cycle, as well as discussing the pathophysiological consequences of the breakdown in this GABAAR regulation.

Pregnancy

One of the most common physiological conditions associated with elevated steroid hormone levels is pregnancy. During pregnancy, progesterone levels rise approximately 200-fold (Backstrom et al., 2003) and, in addition, there are also large increases in the levels of the neuroactive steroids allopregnanolone and THDOC (Concas et al., 1998). Many studies investigating the changes in GABAAR subunit expression related to pregnancy have relied on “pseudopregnancy” or progesterone withdrawal models. Progesterone withdrawal has been shown to increase expression of GABAAR α4 and δ expression in the hippocampus (Smith et al., 1998) and periaqueductal grey (PAG) (Griffiths and Lovick, 2005a). In addition, many studies have focused on changes in mRNA levels, which may not reflect changes in functional protein levels. GABAAR γ2 mRNA levels have been shown to be decreased during pregnancy in the cortex and hippocampus (Concas et al., 1998;Follesa et al., 1998) with no change in the levels of α1, α2, α3, α4, β1, β2, β3 (Concas et al., 1998;Follesa et al., 1998). These changes were blocked by finasteride treatment (Concas et al., 1998), suggesting that this regulation of GABAARs is mediated by neurosteroids.

In order to investigate changes in GABAARs associated with the highly elevated steroid hormone levels throughout pregnancy and postpartum in C57Bl/6 mice, we measured GABAAR subunit expression by Western blot analysis in the hippocampal membrane fraction to ensure only membrane-associated, functional receptors were analyzed. We demonstrated a decreased expression of the GABAAR δ subunit expression in the hippocampus at day 18 of pregnancy, which rebounds to virgin levels by 48 hours postpartum (Table 1) (Maguire and Mody, 2008). The functional consequence of this decrease in the GABAAR δ subunit during pregnancy was analyzed by whole-cell patch clamp recording and corresponds to a decrease in the tonic inhibition mediated by these receptors in dentate gyrus granule cells (Table 1) (Maguire and Mody, 2008). Similarly, we detected a decrease in the GABAAR γ2 subunit in the hippocampus during pregnancy which corresponded to a decrease in the amplitude of spontaneous inhibitory postsynaptic potentials (sIPSCs) measured by whole-cell patch clamp recording, mediated predominantly by these receptors in dentate gyrus granule cells (Table 1) (Maguire and Mody, 2008). These alterations in GABAARs may be compensatory changes in response to the elevated levels of allopregnanolone throughout pregnancy, which become elevated to nearly 100 nM during pregnancy (Paul and Purdy, 1992), within the concentration range which causes sedation (80 – 160 nM) under normal conditions (Sundstrom et al., 1999). The rapid return of neurosteroid levels to pre-pregnancy levels is accompanied by a quick postpartum reversion of GABAAR expression and inhibition to control levels, likely to maintain an ideal level of inhibition throughout the postpartum period (Figure 1). Our data suggest that GABAARs, which are sensitive to the high levels of neurosteroids which occur throughout pregnancy, likely become downregulated to prevent excess inhibition throughout pregnancy which may have adverse behavioral consequences. However, recently it was discovered that these same receptors undergo a different pattern of regulation throughout pregnancy in the rat (Sanna et al., 2009), suggesting that the regulation of GABAARs throughout pregnancy may be more complex than merely homeostatic plasticity and that there may also be species differences in steroid hormone-mediated GABAAR regulation.

Table 1. Steroid Hormone-Associated Changes in GABAAR Subunit Expression and Function.

Western blot analysis was performed on the total hippocampal membrane fraction and probed with an antibody for the GABAAR δ subunit or the GABAAR γ2 subunit. Optical density measurements are shown in Table 1 (left; Maguire, 2005 and Maguire, 2008). Altered GABAAR subunit expression is associated with changes in GABAergic inhibition. The tonic GABAergic inhibition and peak amplitude of sIPSCs recorded in dentate gyrus granule cells is shown in Table 1 (right; Maguire, 2005 and Maguire, 2008).

δ Expression γ 2 Expression Tonic Inhibition Phasic Inhibition

Optical Density (a.u.) Tonic Current (pA) Peak Amplitude (pA)
Ovarian Cycle
Estrus 0.37 +/- 0.04 0.51 +/- 0.02 29.8 +/- 5.5 90.6 +/- 13.4
Diestrus 0.53 +/- 0.01 * 0.34 +/- 0.01 * 57.6 +/- 10.2 * 114.5 +/- 20.5

Pregnancy
Virgin 0.42 +/- 0.03 0.55 +/- 0.01 39.8 +/- 4.4 76.4 +/- 5.5
Pregnancy 0.28 +/- 0.02 * 0.37 +/- 0.01 * 19.7 +/- 3.6 * 45.4 +/- 3.9 *
Postpartum 0.42 +/- 0.05 0.55 +/- 0.01 40.4 +/- 7.1 69.5 +/- 6.9
*

denotes p < 0.05 using a one-way ANOVA. Estrous cycle and Pregnancy data were analyzed separately, Maguire, 2005 and Maguire, 2008, respectively.

Figure 1. A Model of GABAAR Dysfunction in Postpartum Depression.

Figure 1

In the face of elevated steroid hormone levels during late pregnancy, the neurosteroid sensitive GABAAR δ and γ2 subunits must become downregulated to maintain a level of inhibition throughout pregnancy. At the time of parturition, as steroid hormone levels rapidly decline, the levels of GABAAR δ and γ2 subunits must be recovered to pre-pregnancy levels to maintain an ideal level of inhibition throughout the postpartum period. Inability to regulate GABAARs during the postpartum period (dotted line) may result in an imbalance between excitation and inhibition resulting in abnormal postpartum mood disorders, such as postpartum depression.

We propose that a deficiency in GABAAR regulation throughout pregnancy and postpartum may predispose individuals to mood disorders associated with the postpartum period, such as postpartum depression (Figure 1). The postpartum period is a particularly vulnerable period for mood disorders and this vulnerability is thought to be related to changing steroid hormone levels. However, it may not be the steroid hormones per se but rather the site of steroid hormone action in the CNS, namely the GABAAR δ subunit. Consistent with this hypothesis, the inability to properly regulate GABAARs during pregnancy and postpartum, as in mice that are deficient in the GABAAR δ subunit (Gabrd−/− mice), is associated with depression-like and abnormal maternal behaviors in these mice. Gabrd−/− mice exhibit depression-like behaviors 48 hours postpartum assessed using the Porsolt forced swim test. In addition, postpartum Gabrd−/− mice fail to build a nest, they keep their pups at an increased distance from the mother, and exhibit an increased in the percentage of pups that die due to neglect and/or cannibalism (Maguire and Mody, 2008). Our data are consistent with the idea that the pathophysiology of postpartum depression may be related to the inability to properly regulate the expression of GABAARs throughout pregnancy and restore their function postpartum (Figure 1).

Stress

Another physiological condition known to elevate steroid hormone levels is stress. Stress activates the hypothalamic-pituitary-adrenal axis resulting in the release of corticosteroids, thus, resulting in an increase in both circulating and brain levels of the neuroactive steroid THDOC and allopregnanolone (Mcewen, 2002;Purdy et al., 1991;Reddy and Rogawski, 2002). Acute stress has been shown to increase THDOC levels (Barbaccia et al., 1996a;Barbaccia et al., 2001) from 1-5 nM to 15-30 nM (Reddy and Rogawski, 2002); for review see (Reddy, 2003)) and allopregnanolone levels 8-fold (Purdy et al., 1991). However, stress-related neurosteroids exert complex actions on GABAARs. Several studies, relying heavily on binding and uptake assays, have demonstrated changes in GABAAR function following stress (Akinci and Johnston, 1993;Schwartz et al., 1987;Serra et al., 2000;Skerritt et al., 1981). For instance, acute stress has been shown to increase GABA receptor-mediated chloride influx (Schwartz et al., 1987), while a decrease in GABAergic function is associated with chronic stress (Serra et al., 2000). An acute swim stress enhances the binding of GABA agonists (Akinci and Johnston, 1993;Skerritt et al., 1981) and increases the seizure threshold induced by GABA antagonists (Pericic et al., 2001). It has been suggested that elevated THDOC levels following acute stress are responsible for the decreased seizure susceptibility (Reddy, 2003), such that the decreased seizure susceptibility is correlated with a 3-fold increase in circulating levels of THDOC (Reddy and Rogawski, 2002). However, elevated levels of neurosteroids following stress induce alterations in GABAARs which may also contribute to altered neuronal excitability.

We demonstrate alterations in GABAAR subunit expression following an acute stressful episode, which may function to maintain the balance between excitation and inhibition following stress. Mice subjected to a 2 min CO2 stress exhibit an increase in GABAAR δ subunit expression 30 mins following the acute stressor (Maguire and Mody, 2007), which is associated with an increase in tonic GABAergic inhibition in dentate gyrus granule cells assessed by whole-cell patch clamp recording (Maguire and Mody, 2007). Previous binding studies have suggested that there is a decrease in GABAergic function 10 mins following an acute stressful episodes and that the increase in neurosteroid levels at 30 mins following the stressor may serve to restore GABAergic function following stress (Barbaccia et al., 1996b). The dynamic regulation of GABAARs in response to an acute stressor is a compensatory mechanism to cope with the stress and disruption in the regulation of GABAARs in response to stress may underlie the stress-induced exacerbation of many psychiatric and neurological disorders.

Ovarian Cycle

Changes in neuronal excitability and anxiety are associated with changes in steroid hormone levels over the ovarian cycle (Backstrom et al., 2003;Herzog et al., 1997;Herzog et al., 2004), which have been attributed to changes in endogenous neurosteroid levels (Backstrom et al., 2003;Herzog et al., 1997;Herzog et al., 2004). However, how neurosteroids alter neuronal excitability over the ovarian cycle is unclear, but is thought to involve actions of neurosteroids on GABAARs. Changes in GABAergic inhibition over the ovarian cycle have been inferred from alterations in muscimol binding and changes in the sensitivity to neurosteroids and benzodiazepines over the estrous cycle (Aldahan et al., 1994;Martin and Williams, 1995;Molina-Hernandez and Tellez-Alcantara, 2001;Molina-Hernandez et al., 2001;Reddy and Kulkarni, 1999;Sundstrom et al., 1998;Aldahan et al., 1994;Martin and Williams, 1995;Molina-Hernandez and Tellez-Alcantara, 2001;Molina-Hernandez et al., 2001;Sundstrom et al., 1998;Reddy and Kulkarni, 1999). In addition to the direct effects of neurosteroids in potentiating the effects of GABA on GABAARs, steroid hormones have also been shown to alter GABAAR subunit composition. For example, short-term exposure to progesterone or allopregnanolone increases GABAAR α4 and δ subunit expression in the hippocampus, with peak expression at 2-3 days of treatment (Gulinello et al., 2001;Hsu and Smith, 2003). Direct changes in GABAARs over the estrous cycle of mice have been reported in the hippocampus (Maguire et al., 2005) as well as in the periaqueductal grey (PAG) (Griffiths and Lovick, 2005a;Griffiths and Lovick, 2005b). Our data demonstrate an increase in the expression of the GABAAR δ subunit and a decrease in GABAAR γ2 δ subunit by Western blot analysis at times of the estrous cycle in mice characterized by elevated levels of the steroid hormone, progesterone (Table 1) (Maguire et al., 2005). These changes in GABAAR subunit expression are correlated with elevated levels of progesterone (Maguire et al., 2005), and although the levels of allopregnanolone were not directly measured in this study, allopregnanolone levels have been shown to mirror changes in progesterone levels (in review (Paul and Purdy, 1992)). The increase in the GABAAR δ subunit expression over the ovarian cycle is associated with an increase in the tonic inhibition measured by whole-cell patch clamp recording mediated by these receptors in dentate gyrus granule cells (Table 1). However, we do not see a change in the phasic component of GABAergic inhibition despite the decreased levels of GABAAR γ2 subunit expression. This is likely due to the fact that the synaptic receptors are saturated upon release of GABA or these receptors sit peri- or extrasynaptically where they would not sense synaptically released GABA. These changes in GABAAR structure and function over the ovarian cycle are associated with a decrease is seizure susceptibility, and a decrease in anxiety levels (Maguire et al., 2005). These data suggest that changes in neuronal excitability over the ovarian cycle may involve regulation of GABAARs and dysregulation may play a role in PMS and/or PMDD.

Significance

Elucidating the neurosteroid-mediated alterations in GABAAR composition has altered the way that scientists think about steroid hormone-associated neuropsychiatric disorders. Previously, it was thought that steroid hormones acted solely through steroid hormone receptors to induce changes in gene transcription and that steroid hormone derivatives, or neurosteroids, exerted only acute effects on GABAARs. It is now clear that steroid hormones can alter GABAAR subunit composition and thereby alter GABAergic inhibition, independent of steroid hormone receptors (Maguire and Mody, 2007). Interestingly, acute exposure to steroid hormones/neurosteroids have a different effect than prolonged exposure to these same compounds, highlighting the bimodal effects of steroid hormones and their derivatives. For example, in response to a modest, brief increase in neurosteroid levels, such as over the ovarian cycle or following acute stress, there is an increase in the tonic inhibition with no change in the phasic component of inhibition. In contrast, in response to very high levels of neurosteroids for a prolonged period of time, such as during pregnancy, both the tonic and phasic components of inhibition become dampened. These dose-dependent changes in inhibition may be related to differences in the affinity of different receptors assemblies for neurosteroids. At low nanomolar concentrations, neurosteroids act preferentially at δ subunit containing receptors which mediate the phasic component of inhibition. However, at higher concentrations neurosteroids can also activate γ2 subunit-containing receptors. Therefore, the concentration and time of exposure of neurosteroids may have an effect GABAAR regulation. These alterations in GABAARs likely play an important role in steroid hormone-associated changes in neuronal excitability and dysfunction in the regulation of GABAARs by steroid hormones may play a role in steroid hormone-associated neuropsychiatric disorders (Longone et al., 2008;Girdler and Klatzkin, 2007), such as PMS, PMDD, and postpartum depression (Backstrom et al., 2003;Sundstrom et al., 1999).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Reference List

  1. Akinci MK, Johnston GA. Sex differences in acute swim stress-induced changes in the binding of MK-801 to the NMDA subclass of glutamate receptors in mouse forebrain. J Neurochem. 1993;61:2290–2293. doi: 10.1111/j.1471-4159.1993.tb07472.x. [DOI] [PubMed] [Google Scholar]
  2. Aldahan MI, Tehrani MHJ, Thalmann RH. Regulation of Gamma-Aminobutyric Acid(B) (Gaba(B)) Receptors in Cerebral-Cortex During the Estrous-Cycle. Brain Research. 1994;640:33–39. doi: 10.1016/0006-8993(94)91854-6. [DOI] [PubMed] [Google Scholar]
  3. Backstrom T, et al. Pathogenesis in menstrual cycle-linked CNS disorders. Steroids and the Nervous System. 2003;1007:42–53. doi: 10.1196/annals.1286.005. [DOI] [PubMed] [Google Scholar]
  4. Barbaccia ML, Concas A, Roscetti G, Bolacchi F, Mostallino MC, Purdy RH, Biggio G. Stress-induced increase in brain neuroactive steroids: Antagonism by abecarnil. Pharmacology Biochemistry and Behavior. 1996a;54:205–210. doi: 10.1016/0091-3057(95)02133-7. [DOI] [PubMed] [Google Scholar]
  5. Barbaccia ML, Roscetti G, Trabucchi M, Mostallino MC, Concas A, Purdy RH, Biggio G. Time-dependent changes in rat brain neuroactive steroid concentrations and GABA(A) receptor function after acute stress. Neuroendocrinology. 1996b;63:166–172. doi: 10.1159/000126953. [DOI] [PubMed] [Google Scholar]
  6. Barbaccia ML, Serra M, Purdy RH, Biggio G. Stress and neuroactive steroids. Neurosteroids and Brain Function. 2001;46:243–272. doi: 10.1016/s0074-7742(01)46065-x. [DOI] [PubMed] [Google Scholar]
  7. Belelli D, Casula A, Ling A, Lambert JJ. The influence of subunit composition on the interaction of neurosteroids with GABA(A) receptors. Neuropharmacology. 2002;43:651–661. doi: 10.1016/s0028-3908(02)00172-7. [DOI] [PubMed] [Google Scholar]
  8. Belelli D, Lambert JJ. Neurosteroids: endogenous regulators of the GABA(A) receptor. Nat Rev Neurosci. 2005;6:565–575. doi: 10.1038/nrn1703. [DOI] [PubMed] [Google Scholar]
  9. Bloch M, Schmidt PJ, Danaceau M, Murphy J, Nieman L, Rubinow DR. Effects of gonadal steroids in women with a history of postpartum depression. American Journal of Psychiatry. 2000;157:924–930. doi: 10.1176/appi.ajp.157.6.924. [DOI] [PubMed] [Google Scholar]
  10. Bremner JD, Innis RB, White T, Fujita M, Silbersweig D, Goddard AW, Staib L, Stern E, Cappiello A, Woods S, Baldwin R, Charney DS. SPECT [I-123]Iomazenil measurement of the benzodiazepine receptor in panic disorder. Biological Psychiatry. 2000;47:96–106. doi: 10.1016/s0006-3223(99)00188-2. [DOI] [PubMed] [Google Scholar]
  11. Brown N, Kerby J, Bonnert TP, Whiting PJ, Wafford KA. Pharmacological characterization of a novel cell line expressing human alpha(4)beta(3)delta GABA(A) receptors. British Journal of Pharmacology. 2002;136:965–974. doi: 10.1038/sj.bjp.0704795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Concas A, Mostallino MC, Porcu P, Follesa P, Barbaccia ML, Trabucchi M, Purdy RH, Grisenti P, Biggio G. Role of brain allopregnanolone in the plasticity of gamma-aminobutyric acid type A receptor in rat brain during pregnancy and after delivery. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:13284–13289. doi: 10.1073/pnas.95.22.13284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Farrant M, Nusser Z. Variations on an inhibitory theme: Phasic and tonic activation of GABA(A) receptors. Nature Reviews Neuroscience. 2005;6:215–229. doi: 10.1038/nrn1625. [DOI] [PubMed] [Google Scholar]
  14. Follesa P, Floris S, Tuligi G, Mostallino MC, Concas A, Biggio G. Molecular and functional adaptation of the GABA(A) receptor complex during pregnancy and after delivery in the rat brain. European Journal of Neuroscience. 1998;10:2905–2912. doi: 10.1111/j.1460-9568.1998.00300.x. [DOI] [PubMed] [Google Scholar]
  15. Girdler SS, Klatzkin R. Neurosteroids in the context of stress: implications for depressive disorders. Pharmacol Ther. 2007;116:125–139. doi: 10.1016/j.pharmthera.2007.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Goddard AW, Mason GF, Almai A, Rothman DL, Behar KL, Petroff OAC, Charney DS, Krystal JH. Reductions in occipital cortex GABA levels in panic disorder detected with H-1-magnetic resonance spectroscopy. Archives of General Psychiatry. 2001;58:556–561. doi: 10.1001/archpsyc.58.6.556. [DOI] [PubMed] [Google Scholar]
  17. Griffiths J, Lovick T. Withdrawal from progesterone increases expression of alpha4, beta1, and delta GABA(A) receptor subunits in neurons in the periaqueductal gray matter in female Wistar rats. J Comp Neurol. 2005a;486:89–97. doi: 10.1002/cne.20540. [DOI] [PubMed] [Google Scholar]
  18. Griffiths JL, Lovick TA. GABAergic neurones in the rat periaqueductal grey matter express alpha4, beta1 and delta GABAA receptor subunits: plasticity of expression during the estrous cycle. Neuroscience. 2005b;136:457–466. doi: 10.1016/j.neuroscience.2005.08.013. [DOI] [PubMed] [Google Scholar]
  19. Gulinello M, Gong QH, Li X, Smith SS. Short-term exposure to a neuroactive steroid increases alpha 4 GABA(A) receptor subunit levels in association with increased anxiety in the famale rat. Brain Research. 2001;910:55–66. doi: 10.1016/s0006-8993(01)02565-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Herd MB, Belelli D, Lambert JJ. Neurosteroid modulation of synaptic and extrasynaptic GABA(A) receptors. Pharmacol Ther. 2007;116:20–34. doi: 10.1016/j.pharmthera.2007.03.007. [DOI] [PubMed] [Google Scholar]
  21. Herzog AG, Harden CL, Liporace J, Pennell P, Schomer DL, Sperling M, Fowler K, Nikolov B, Shuman S, Newman M. Frequency of catamenial seizure exacerbation in women with localization-related epilepsy. Annals of Neurology. 2004;56:431–434. doi: 10.1002/ana.20214. [DOI] [PubMed] [Google Scholar]
  22. Herzog AG, Klein P, Ransil BJ. Three patterns of catamenial epilepsy. Epilepsia. 1997;38:1082–1088. doi: 10.1111/j.1528-1157.1997.tb01197.x. [DOI] [PubMed] [Google Scholar]
  23. Hevers W, Luddens H. The diversity of GABAA receptors - Pharmacological and electrophysiological properties of GABAA channel subtypes. Molecular Neurobiology. 1998;18:35–86. doi: 10.1007/BF02741459. [DOI] [PubMed] [Google Scholar]
  24. Hosie AM, Clarke L, da SH, Smart TG. Conserved site for neurosteroid modulation of GABA A receptors. Neuropharmacology. 2009;56:149–154. doi: 10.1016/j.neuropharm.2008.07.050. [DOI] [PubMed] [Google Scholar]
  25. Hosie AM, Wilkins ME, da Silva HM, Smart TG. Endogenous neurosteroids regulate GABAA receptors through two discrete transmembrane sites. Nature. 2006;444:486–489. doi: 10.1038/nature05324. [DOI] [PubMed] [Google Scholar]
  26. Hsu FC, Smith SS. Progesterone withdrawal reduces paired-pulse inhibition in rat hippocampus: Dependence on GABA(A) receptor alpha 4 subunit upregulation. Journal of Neurophysiology. 2003;89:186–198. doi: 10.1152/jn.00195.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Longone P, Rupprecht R, Manieri GA, Bernardi G, Romeo E, Pasini A. The complex roles of neurosteroids in depression and anxiety disorders. Neurochem Int. 2008;52:596–601. doi: 10.1016/j.neuint.2007.10.001. [DOI] [PubMed] [Google Scholar]
  28. Maguire J, Mody I. GABA(A)R plasticity during pregnancy: relevance to postpartum depression. Neuron. 2008;59:207–213. doi: 10.1016/j.neuron.2008.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Maguire J, Mody I. Neurosteroid synthesis-mediated regulation of GABA(A) receptors: relevance to the ovarian cycle and stress. J Neurosci. 2007;27:2155–2162. doi: 10.1523/JNEUROSCI.4945-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Maguire JL, Stell BM, Rafizadeh M, Mody I. Ovarian cycle-linked changes in GABA(A) receptors mediating tonic inhibition alter seizure susceptibility and anxiety. Nat Neurosci. 2005;8(6):797–804. doi: 10.1038/nn1469. [DOI] [PubMed] [Google Scholar]
  31. Malizia AL, Cunningham VJ, Bell CJ, Liddle PF, Jones T, Nutt DJ. Decreased brain GABA(A)-benzodiazepine receptor binding in panic disorder - Preliminary results from a quantitative PET study. Archives of General Psychiatry. 1998;55:715–720. doi: 10.1001/archpsyc.55.8.715. [DOI] [PubMed] [Google Scholar]
  32. Martin JV, Williams DB. Benzodiazepine Binding Varies with Stage of Estrous-Cycle in Unwashed Membranes from Mouse-Brain. Life Sciences. 1995;57:1903–1909. doi: 10.1016/0024-3205(95)02177-k. [DOI] [PubMed] [Google Scholar]
  33. Mcewen BS. The neurobiology and neuroendocrinology of stress Implications for post-traumatic stress disorder from a basic science perspective. Psychiatric Clinics of North America. 2002;25:469. doi: 10.1016/s0193-953x(01)00009-0. [DOI] [PubMed] [Google Scholar]
  34. Mihalek RM, Banerjee PK, Korpi ER, Quinlan JJ, Firestone LL, Mi ZP, Lagenaur C, Tretter V, Sieghart W, Anagnostaras SG, Sage JR, Fanselow MS, Guidotti A, Spigelman I, Li ZW, DeLorey TM, Olsen RW, Homanics GE. Attenuated sensitivity to neuroactive steroids in gamma-aminobutyrate type A receptor delta subunit knockout mice. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:12905–12910. doi: 10.1073/pnas.96.22.12905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Mitchell EA, Herd MB, Gunn BG, Lambert JJ, Belelli D. Neurosteroid modulation of GABAA receptors: molecular determinants and significance in health and disease. Neurochem Int. 2008;52:588–595. doi: 10.1016/j.neuint.2007.10.007. [DOI] [PubMed] [Google Scholar]
  36. Mody I, Pearce RA. Diversity of inhibitory neurotransmission through GABAA receptors. Trends in Neurosciences. 2004;27:569–575. doi: 10.1016/j.tins.2004.07.002. [DOI] [PubMed] [Google Scholar]
  37. Molina-Hernandez M, Contreras CM, Tellez-Alcantara P. Diazepam increases the number of punished responses in a conflict-operant paradigm during late proestrus and estrus in the Wistar rat. Neuropsychobiology. 2001;43:29–33. doi: 10.1159/000054862. [DOI] [PubMed] [Google Scholar]
  38. Molina-Hernandez M, Tellez-Alcantara NP. Estrus variation in anticonflict effects of midazolam microinjected into septal nuclei in female Wistar rats. Pharmacology Biochemistry and Behavior. 2001;68:531–537. doi: 10.1016/s0091-3057(01)00459-2. [DOI] [PubMed] [Google Scholar]
  39. Nusser Z, Sieghart W, Somogyi P. Segregation of different GABA(A) receptors to synaptic and extrasynaptic membranes of cerebellar granule cells. Journal of Neuroscience. 1998;18:1693–1703. doi: 10.1523/JNEUROSCI.18-05-01693.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Olsen RW, Sieghart W. International Union of Pharmacology. LXX. Subtypes of gamma-aminobutyric acid(A) receptors: classification on the basis of subunit composition, pharmacology, and function Update. Pharmacol Rev. 2008;60:243–260. doi: 10.1124/pr.108.00505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Paul SM, Purdy RH. Neuroactive Steroids. Faseb Journal. 1992;6:2311–2322. [PubMed] [Google Scholar]
  42. Peng Z, Hauer B, Mihalek RM, Homanics GE, Sieghart W, Olsen RW, Houser CR. GABA(A) receptor changes in delta subunit-deficient mice: Altered expression of alpha 4 and gamma 2 subunits in the forebrain. Journal of Comparative Neurology. 2002;446:179–197. doi: 10.1002/cne.10210. [DOI] [PubMed] [Google Scholar]
  43. Peng ZC, Huang CS, Stell BM, Mody I, Houser CR. Altered expression of the delta subunit of the GABA(A) receptor in a mouse model of temporal lobe epilepsy. Journal of Neuroscience. 2004;24:8629–8639. doi: 10.1523/JNEUROSCI.2877-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Pericic D, Jazvinscak M, Svob D, Mirkovic K. Swim stress alters the behavioural response of mice to GABA-related and some GABA-unrelated convulsants. Epilepsy Res. 2001;43:145–152. doi: 10.1016/s0920-1211(00)00194-7. [DOI] [PubMed] [Google Scholar]
  45. Petursson H. The Benzodiazepine Withdrawal Syndrome. Addiction. 1994;89:1455–1459. doi: 10.1111/j.1360-0443.1994.tb03743.x. [DOI] [PubMed] [Google Scholar]
  46. Pirker S, Schwarzer C, Wieselthaler A, Sieghart W, Sperk G. GABA(A) receptors: Immunocytochemical distribution of 13 subunits in the adult rat brain. Neuroscience. 2000;101:815–850. doi: 10.1016/s0306-4522(00)00442-5. [DOI] [PubMed] [Google Scholar]
  47. Purdy RH, Morrow AL, Moore PH, Paul SM. Stress-Induced Elevations of Gamma-Aminobutyric-Acid Type-A Receptor-Active Steroids in the Rat-Brain. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:4553–4557. doi: 10.1073/pnas.88.10.4553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Reddy DS. Is there a physiological role for the neurosteroid THDOC in stress-sensitive conditions? Trends in Pharmacological Sciences. 2003;24:103–106. doi: 10.1016/S0165-6147(03)00023-3. [DOI] [PubMed] [Google Scholar]
  49. Reddy DS, Kulkarni SK. Sex and estrous cycle-dependent changes in neurosteroid and benzodiazepine effects on food consumption and plus-maze learning behaviors in rats. Pharmacology Biochemistry and Behavior. 1999;62:53–60. doi: 10.1016/s0091-3057(98)00126-9. [DOI] [PubMed] [Google Scholar]
  50. Reddy DS, Rogawski MA. Stress-induced deoxycorticosterone-derived neurosteroids modulate GABA(A) receptor function and seizure susceptibility. Journal of Neuroscience. 2002;22:3795–3805. doi: 10.1523/JNEUROSCI.22-09-03795.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Sanna E, Mostallino MC, Murru L, Carta M, Talani G, Zucca S, Mura ML, Maciocco E, Biggio G. Changes in Expression and Function of Extrasynaptic GABAA Receptors in the Rat Hippocampus during Pregnancy and after Delivery. J Neurosci. 2009;29:1755–1765. doi: 10.1523/JNEUROSCI.3684-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Schwartz RD, Wess MJ, Labarca R, Skolnick P, Paul SM. Acute Stress Enhances the Activity of the Gaba Receptor-Gated Chloride-Ion Channel in Brain. Brain Research. 1987;411:151–155. doi: 10.1016/0006-8993(87)90692-5. [DOI] [PubMed] [Google Scholar]
  53. Serra M, Pisu MG, Littera M, Papi G, Sanna E, Tuveri F, Usala L, Purdy RH, Biggio G. Social isolation-induced decreases in both the abundance of neuroactive steroids and GABA(A) receptor function in rat brain. Journal of Neurochemistry. 2000;75:732–740. doi: 10.1046/j.1471-4159.2000.0750732.x. [DOI] [PubMed] [Google Scholar]
  54. Skerritt JH, Trisdikoon P, Johnston GA. Increased GABA binding in mouse brain following acute swim stress. Brain Res. 1981;215:398–403. doi: 10.1016/0006-8993(81)90524-2. [DOI] [PubMed] [Google Scholar]
  55. Smith MJ, Adams LF, Schmidt PJ, Rubinow DR, Wassermann EM. Abnormal luteal phase excitability of the motor cortex in women with premenstrual syndrome. Biological Psychiatry. 2003;54:757–762. doi: 10.1016/s0006-3223(02)01924-8. [DOI] [PubMed] [Google Scholar]
  56. Smith SS, Gong QH, Hsu FC, Markowitz RS, ffrench-Mullen JMH, Li HS. GABA(A) receptor alpha 4 subunit suppression prevents withdrawal properties of an endogenous steroid. Nature. 1998;392:926–930. doi: 10.1038/31948. [DOI] [PubMed] [Google Scholar]
  57. Smith SS, Shen H, Gong QH, Zhou X. Neurosteroid regulation of GABA(A) receptors: Focus on the alpha4 and delta subunits. Pharmacol Ther. 2007;116:58–76. doi: 10.1016/j.pharmthera.2007.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Spigelman I, Li ZW, Liang J, Cagetti E, Samzadeh S, Mihalek RM, Homanics GE, Olsen RW. Reduced inhibition and sensitivity to neurosteroids in hippocampus of mice lacking the GABA(A) receptor delta subunit. Journal of Neurophysiology. 2003;90:903–910. doi: 10.1152/jn.01022.2002. [DOI] [PubMed] [Google Scholar]
  59. Stell BM, Brickley SG, Tang CY, Farrant M, Mody I. Neuroactive steroids reduce neuronal excitability by selectively enhancing tonic inhibition mediated by delta subunit-containing GABAA receptors. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:14439–14444. doi: 10.1073/pnas.2435457100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Stoffel-Wagner B. Neurosteroid metabolism in the human brain. Eur J Endocrinol. 2001;145:669–679. doi: 10.1530/eje.0.1450669. [DOI] [PubMed] [Google Scholar]
  61. Strohle A, Romeo E, di Michele F, Pasini A, Yassouridis A, Holsboer F, Rupprecht R. GABA(A) receptor-modulating neuroactive steroid composition in patients with panic disorder before and during paroxetine treatment. American Journal of Psychiatry. 2002;159:145–147. doi: 10.1176/appi.ajp.159.1.145. [DOI] [PubMed] [Google Scholar]
  62. Sundstrom I, Andersson A, Nyberg S, Ashbrook D, Purdy RH, Backstrom T. Patients with premenstrual syndrome have a different sensitivity to a neuroactive steroid during the menstrual cycle compared to control subjects. Neuroendocrinology. 1998;67:126–138. doi: 10.1159/000054307. [DOI] [PubMed] [Google Scholar]
  63. Sundstrom I, Ashbrook D, Backstrom T. Reduced benzodiazepine sensitivity in patients with premenstrual syndrome: A pilot study. Psychoneuroendocrinology. 1997;22:25–38. doi: 10.1016/s0306-4530(96)00035-2. [DOI] [PubMed] [Google Scholar]
  64. Sundstrom I, Backstrom T, Wang M, Olsson T, Seippel L, Bixo M. Premenstrual syndrome, neuroactive steroids and the brain. Gynecological Endocrinology. 1999;13:206–220. doi: 10.3109/09513599909167557. [DOI] [PubMed] [Google Scholar]
  65. Tiihonen J, Kuikka J, Rasanen P, Lepola U, Koponen H, Liuska A, Lehmusvaara A, Vainio P, Kononen M, Bergstrom K, Yu M, Kinnunen I, Akerman K, Karhu J. Cerebral benzodiazepine receptor binding and distribution in generalized anxiety disorder: a fractal analysis. Molecular Psychiatry. 1997;2:463–471. doi: 10.1038/sj.mp.4000329. [DOI] [PubMed] [Google Scholar]
  66. Walker MC, Semyanov A. Regulation of excitability by extrasynaptic GABA(A) receptors. Results Probl Cell Differ. 2008;44:29–48. 29–48. doi: 10.1007/400_2007_030. [DOI] [PubMed] [Google Scholar]
  67. Wei WZ, Zhang NH, Peng ZC, Houser CR, Mody I. Perisynaptic localization of delta subunit-containing GABAA receptors and their activation by GABA spillover in the mouse dentate gyrus. Journal of Neuroscience. 2003;23:10650–10661. doi: 10.1523/JNEUROSCI.23-33-10650.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Whiting PJ, Bonnert TP, McKernan RM, Farrar S, Le Bourdelles B, Heavens RP, Smith DW, Hewson L, Rigby MR, Sirinathsinghji DJS, Thompson SA, Wafford KA. Molecular and functional diversity of the expanding GABA-A receptor gene family. Molecular and Functional Diversity of Ion Channels and Receptors. 1999;868:645–653. doi: 10.1111/j.1749-6632.1999.tb11341.x. [DOI] [PubMed] [Google Scholar]
  69. Wisden W, Laurie DJ, Monyer H, Seeburg PH. The Distribution of 13-Gaba-A Receptor Subunit Messenger-Rnas in the Rat-Brain .1. Telencephalon, Diencephalon, Mesencephalon. Journal of Neuroscience. 1992;12:1040–1062. doi: 10.1523/JNEUROSCI.12-03-01040.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Wisner KL, Parry BL, Piontek CM. Postpartum depression. New England Journal of Medicine. 2002;347:194–199. doi: 10.1056/NEJMcp011542. [DOI] [PubMed] [Google Scholar]
  71. Wohlfarth KM, Bianchi MT, Macdonald RL. Enhanced neurosteroid potentiation of ternary GABA(A) receptors containing the delta subunit. Journal of Neuroscience. 2002;22:1541–1549. doi: 10.1523/JNEUROSCI.22-05-01541.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES