Open access

Neurosteroid Biosynthesis Upregulation: A Novel Promising Therapy for Anxiety Disorders and PTSD

Written By

Graziano Pinna

Submitted: 27 October 2010 Published: 01 August 2011

DOI: 10.5772/18472

From the Edited Volume

Anxiety Disorders

Edited by Vladimir Kalinin

Chapter metrics overview

4,617 Chapter Downloads

View Full Metrics

1. Introduction

Generalized anxiety, panic, and posttraumatic stress disorder (PTSD) are debilitating conditions, which have an incidence of one in ten persons in the general population and epidemiological studies also report that these disorders often occur with depression (1-3). Anxiolytic benzodiazepines, including diazepam and alprazolam, remain the best and most used treatments for these conditions (4-7). However, their therapeutic use is associated with side effects, which include sedation and rapid development of tolerance as well as dependence. This results in severe discontinuation symptoms and often to drug abuse (4-6, 8; 9).

In many patients, including patients with PTSD, the pharmacological effects of these drugs are very weak and there is a large number of non-responders (10-12). This has stimulated drug design that for many decades has focused in the development of new more effective therapies for anxiety disorders (13-15). Novel neuronal biomarkers for the pharmacological targets of the next generation of anxiolytic drugs have been discovered.

The downregulation of neurosteroid biosynthesis has been implicated in the pathophysiology of anxiety and depressive disorders (reviewed in 16). Decreases in cerebrospinal fluid (clinical studies) and brain content (preclinical studies) of the GABAA receptor-active progesterone derivative, allopregnanolone, have been associated with affective and mood disorders, which includes depression, anxiety spectrum disorders, PTSD, premenstrual dysphoric disorder, schizophrenia, and impulsivity (17-27). Thus, elevating or normalizing the downregulation of brain allopregnanolone levels could be a promising therapeutic strategy for these psychiatric disorders. This prompted investigations to develop new neurosteroidogenic agents to contrast allopregnanolone biosynthesis deficits in anxiety and depression (28-31).

We measured allopregnanolone levels in the cerebrospinal fluid (CSF) of PTSD patients assuming that allopregnanolone levels in the CSF reflect the levels of this neurosteroid in the brain (17). Also, in depressed patients, the concentration of allopregnanolone in the CSF was decreased by about 50-60% of the levels measured in non-psychiatric patients (26). The CSF allopregnanolone level decrease is likely induced by a downregulation of the expression of 5α-reductase type I mRNA in the prefrontal-cortex (area BA9) that we measured in depressed patients and age- and sex-matched non-psychiatric subjects (32). The cortical level of 5α-reductase mRNA in depressed patients was dramatically decreased to about 50% of the levels measured in non-psychiatric comparison subjects, whereas the levels of 5α-reductase mRNA was unchanged in the cerebellum (32). In depressed patients, SSRI treatment with fluoxetine and fluvoxamine normalized the CSF allopregnanolone content (26) in a manner that correlated with the improvement in depressive symptoms. These results were confirmed in studies that determined allopregnanolone or levels of 5α-tetrahydrodeoxycorticosterone, another positive modulator of GABAA receptor function, in the plasma of depressed patients treated with SSRIs (33).

In premenopausal women with PTSD, the CSF allopregnanolone levels were decreased by about 60% and were inversely correlated with PTSD re-experiencing and comorbid depressive symptoms (17). Interestingly, CSF allopregnanolone levels were lowest in those patients with PTSD and comorbid depression. Also, the ratio of allopregnanolone to its steroid precursor, 5α-dihydroprogesterone (5α-DHP), was decreased among the PTSD patients, suggesting the presence of an impairment in the biosynthesis of allopregnanolone from its precursor 5α-DHP (17). These data suggest that the downregulation of brain allopregnanolone levels in PTSD and depressed patients may cause a GABAergic neurotransmission dysfunction, which in turn results in the behavioral symptoms seen in these patients.

Following the finding that fluoxetine and paroxetine and other SSRIs increase the content of allopregnanolone in several rodent brain structures (34), we hypothesized that normalization of brain allopregnanolone levels may underlie the pharmacological effects of the so called “selective serotonin reuptake inhibitors” or SSRIs in mood disorders. To test this hypothesis, we conducted experiments using the socially isolated mouse as an animal model of anxiety disorders and PTSD (16; 35-38). The socially isolated mouse expresses a robust decrease of corticolimbic allopregnanolone levels, which are associated with anxiety-like behaviors, fear, resistance to sedation, and heightened aggression (16; 35; 39). These behavioral deficits can be ameliorated by administration of fluoxetine and other SSRIs that upregulate allopregnanolone levels. Interestingly, fluoxetine’s pharmacological effects resulted to be independent from the ability of this drug to inhibit serotonin reuptake (35; 36).

Our experiments support a selective and novel mechanism whereby SSRIs, acting as selective brain steroidogenic stimulants (SBSSs), increase brain corticolimbic allopregnanolone levels and improve PTSD, anxiety, and depression behavioral symptoms.

Advertisement

2. Neurosteroids modulation of GABAA receptors function

Biosynthesis of neurosteroids in the brain is independent from adrenals, ovaries, and testis (40-44). Neurosteroids are functionally active in modulating gene expression and neurotransmitter systems (45-52). Allopregnanolone exerts pharmacological actions, such as anticonvulsant, anxiolytic, antidepressant, and even sedative-hypnotic (53-60). These pharmacological actions are similar to those elicited by barbiturates and benzodiazepines (52; 61; 62). Allopregnanolone potently (nM affinity), positively, and allosterically modulates the action of GABA at GABAA receptors (45-51). The endogenous physiological relevance of allopregnanolone is substantiated by its facilitation and fine-tuning of the efficacy of direct GABAA receptor activators and positive allosteric modulators of GABA action at GABAA receptors (43; 47; 48; 63). The demonstration that allopregnanolone potentiates GABA responses via two binding sites in the GABAA receptor that, respectively, mediate the potentiation and the direct activation of the GABAA receptor by allopregnanolone has been pivotal in neurosteroid pharmacology (64). Also, GABAA receptors incorporating α4, α6, and δ subunits in combination with γ and β subunits show higher affinity (nM range) for allopregnanolone (45; 46; 51; 64; 65). Relevant for pharmacological strategies to overcome behavioral deficits resulting from GABAA receptor signal transduction deficits, allopregnanolone allosteric modulation of the action of GABA at GABAA receptors is much less selective than that of benzodiazepines, which are relatively inactive at α4- or α6-containing GABAA receptors (4; 45; 46; 66).

Advertisement

3. Neurosteroid biosynthesis in corticolimbic neurons

A study of the neuronal localization of the neurosteroidogenic enzymes, 5α-reductase type I and 3α-hydroxysteroid dehydrogenase (3α-HSD), has recently showed that these enzymes are not expressed in GABAergic cortical interneurons or glial cells (67). Of note, 5α-reductase and 3α-HSD were highly expressed and co-localized in a region-specific way in primary GABAergic and glutamatergic neurons, including pyramidal neurons, granular cells, reticulo-thalamic neurons, medium spiny neurons of the striatum and nucleus accumbens, and Purkinje cells in the cerebellum (67). This suggested that allopregnanolone synthesized in glutamatergic cortical or hippocampal pyramidal neurons or in granular cells of the dentate gyrus may be secreted in: 1) a paracrine manner which would allow allopregnanolone to reach GABAA receptors located in the synaptic membranes of other cortical or hippocampal pyramidal neurons, or 2) an autocrine fashion which would allow allopregnanolone to act locally by binding post-synaptic or extra-synaptic GABAA receptors located on the same dendrites or cell bodies of the cortical or hippocampal pyramidal neuron in which it was produced (67). Alternatively, allopregnanolone might not be released, but may instead diffuse laterally into synaptosome membranes of the cell bodies or dendritic arborization of glutamatergic neurons in which it is produced to attain intracellular access to specific neurosteroid binding sites of GABAA receptors (67; 68). In the amygdala, for example, this would functionally baffle the effects of concomitant excitatory inputs to glutamatergic projection neurons during exposure to unconditioned stress during fear conditioning or to conditioned stressors during extinction.

On the other hand, allopregnanolone produced in primary output GABAergic neurons from the reticular thalamic nucleus may secrete allopregnanolone simultaneously with GABA to concomitantly act at post-synaptic GABAA receptors inserted in glutamatergic thalamocortical neurons (69). Very similarly, allopregnanolone synthesized by striatal medium spiny GABAergic neurons and cerebellar Purkinje cells may activate post-synaptic GABAA receptors located on cell bodies or dendrites of neurons in the deep cerebellar nuclei (67).

The clarification of allopregnanolone site of synthesis and action across several brain regions has been pivotal to our understanding of the possible mechanisms by which allopregnanolone is secreted and acts at GABAA receptors. These studies underscore the functional role of allopregnanolone in fine tuning the strength of GABAergic neurotransmission under physiological conditions and how deficits in allopregnanolone biosynthesis may result in abnormal behavior.

Advertisement

4. Social isolation induces a selective neuron-specific decrease of 5α-reductase in corticolimbic neurons

Exposure of rodents to protracted social isolation stress for 4-8 weeks induces a decrease in allopregnanolone biosynthesis in several corticolimbic structures as a result of a downregulation of the mRNA and protein expression of 5α-reductase type I (35; 70-73; reviewed in 38). Socially isolated mice show a 70% reduction in the synthesis rate of allopregnanolone and 5α-DHP biosynthesis compared to group-housed mice (35; 72).

Allopregnanolone and 5α-DHP are unevenly distributed and expressed in various brain structures (48; 74). The rodent olfactory bulb shows the highest concentrations of 5α-DHP and allopregnanolone followed by the frontal cortex, hippocampus, amygdala, striatum, and cerebellum (74). Interestingly, the largest decrease of 5α-reductase was found in brain regions regulating emotional behavior, including the amygdala and hippocampus, followed by the olfactory bulb and the frontal cortex (74). The expression of 5α-reductase failed to change in the cerebellum and striatum (74; 75). Decreased 5α-reductase was specifically found in cortical pyramidal neurons of layers V-VI, in hippocampal CA3 pyramidal neurons and glutamatergic granular cells of the dentate gyrus, and in the pyramidal-like neurons of the basolateral amygdala (75). However, 5α-reductase fails to change in GABAergic neurons of the reticular thalamic nucleus, central amygdala, cerebellum, and in the medium spiny neurons of the caudatus and putamen (75). In these brain areas, we confirmed that the decrease of 5α-reductase resulted in a reduction of allopregnanolone levels (74; 76; 77). Social isolation failed to change the expression of 3α-HSD, the mRNA expression of diazepam binding inhibitor, and the expression of the 18 kDa translocase protein (TSPO), which is involved in the transport of cholesterol across the inner mitochondrial membrane and activation of neurosteroidogenesis (reviewed in 72). Thus, the downregulation of 5α-reductase appears to be the main factor responsible for the reduction of corticolimbic allopregnanolone levels.

Advertisement

5. GABAergic neurotransmission deficits resulting from allopregnanolone downregulation

Allopregnanolone biosynthesis downregulation as a result of social isolation stress or pharmacological decrease of allopregnanolone induced by inhibiting 5α-reductase with the potent competitive 5α-reductase inhibitor SKF 105,111 decreases GABAergic neurotransmission as demonstrated by reduced loss of righting reflexes induced by GABAA receptor active ligands. The effects of SKF on the muscimol-, pentobarbital-, benzodiazepine-, or alcohol-induced loss of righting reflex loss can be reversed by the systemic or intracerebroventricular administration of allopregnanolone (43; 48). Likewise, social isolation or SKF-induced decrease of allopregnanolone results in facilitation of the seizure activity induced by several drugs that decrease GABAA receptor function, including picrotoxin (63). Administration of allopregnanolone at doses that have virtually no effects on group-housed control mice normalized the increased susceptibility to picrotoxin-induced seizures in SKF-treated or social isolated mice (63). The protracted social isolation or SKF treatment-induced allopregnanolone biosynthesis downregulation appeared to be the primary reason for the GABAA receptor signal transduction deficits observed in these mice. In fact, seizures induced by kainic acid or strychnine in socially isolated mice are similar to those induced by these agents in group housed mice.

Advertisement

6. Behavioral effects induced by allopregnanolone downregulation in corticolimbic areas

The decrease of allopregnanolone biosynthesis in socially isolated mice has been associated with several behavioral deficits that resemble behavioral abnormalities observed in patients with PTSD (16; 17; 30; 38). Hence, this mouse model can be used to study the behavioral responses elicited by treatment with neurosteroidogenic agents, the SBSSs. This new class of drugs includes the SSRI antidepressants that have been shown to elicit a potent neurosteroidogenic activity selectively at low doses as their principal action.

Allopregnanolone has emerged as an important biomarker of emotional behavioral deficits (16; 35-38; 72). This was demonstrated by experiments using socially isolated mice to induce a downregulation of allopregnanolone biosynthesis. We have established a fundamental role for allopregnanolone in the regulation of anxiety-like and aggressive behavior as well as contextual fear conditioning, (16; 37; 63; 74; 77). When mice are socially isolated for a period varying from one to eight weeks, there is a time-dependent increase in aggressive behavior over the first four weeks of isolation, which is inversely correlated with a time-dependent decrease of corticolimbic allopregnanolone levels (35). Likewise, socially isolated mice exposed to a classical fear conditioning paradigm showed enhanced conditioned contextual but not cued fear responses compared with group housed mice (74). The time-related increase of contextual fear responses correlated with the downregulation of 5α-reductase mRNA and protein expression observed in the frontal cortex, hippocampus, and amygdala (74). Socially isolated mice also exhibited impaired and incomplete fear extinction (74). Of note, socially isolated mice also exhibit higher levels of anxiety-like behavior, determined by the elevated plus maze and in the open field (16; 39).

Allopregnanolone plays a pivotal rather than incidental role in the regulation of contextual fear responses and aggression. In fact, pharmacological treatment with allopregnanolone dose-dependently decreased aggression in a manner that correlated with an increase in corticolimbic allopregnanolone content (35). Allopregnanolone also normalized the exaggerated contextual fear responses and anxiety of socially isolated mice (74). Further, administration of the potent 5α-reductase competitive inhibitor SKF 105,111 to normal group-housed mice (43; 48; 47) rapidly (~1 h) decreased levels of allopregnanolone in the olfactory bulb, frontal cortex, hippocampus, and amygdala by 80-90% (73; 74) in association with a dose-dependent increase of conditioned contextual fear responses (74). Administering allopregnanolone doses that normalized hippocampus allopregnanolone levels reversed the effects of SKF 105,111 on conditioned contextual fear responses (74). These results are in agreement with results of many other investigators who have observed that allopregnanolone elicits anxiolytic and antidepressant effects (39; 54; 78-84).

Advertisement

7. Social isolation induces changes in GABAA receptor subunit expression

Postmortem studies suggest that altered corticolimbic GABAergic neurotransmission, GABA receptor binding and receptor subunit composition, as well as GABA synthesis and transport may be associated with various psychiatric disorders, including anxiety disorders, schizophrenia, and depression (85-88).

The regional distribution of GABAA receptor subunit subtypes plays an important role in the pharmacology of GABAA receptor ligands that bind to selective and specific GABAA receptor subunits (89-90). Recent studies showed that α1-containing GABAA receptors mediate the sedative properties of specific GABAergic ligands, such as diazepam, in the same way α2 and probably α3 subunits mediate the anxiolytic effects of benzodiazepines, and α5 subunits appear to be involved in learning and cognition (89; 90). High affinity binding of benzodiazepine to GABAA receptors requires the interaction of α and γ subunits (89; 90).

In socially isolated mice, we found changes in the mRNA and protein expression of several GABAA receptor subunits in the frontal cortex and hippocampus (91). The mRNA levels encoding α1, α2, and γ2 GABAA receptor subunit subtypes were reduced (~50%), while the mRNAs encoding α4 and α5 subunits were increased (~130%) compared to levels measured in group-housed mice (91). Protein levels of α1 and α5 determined in synaptic membrane preparations in the frontal cortex and hippocampus confirmed the former results. Using a laser microdissection technique coupled with nested RT-PCR amplification, we found that α1 mRNA levels were decreased by 50% in layer I neuropil, whereas the expression of α1 subunit mRNA in the pyramidal neurons of layer V was unchanged as a result of social isolation. Thus, changes in GABAA receptor subunits within one brain area are region-specific (91).

Changes in GABAA receptor subunit subtype composition are expected to result in altered pharmacological responses to various GABAA receptor ligands in socially isolated mice. As expected, socially isolated mice showed resistance to the sedative and anxiolytic properties of diazepam and zolpidem, positive allosteric GABAA receptor modulators that bind with high affinity to α1, α2, α3 or α5 subunit-containing GABAA receptors (diazepam) and to α1 subunit-containing GABAA receptors (zolpidem) (91). The α1 subunit of the GABAA receptor plays a primary role in mediating the sedative pharmacological effects of diazepam and zolpidem (92). Hence, their altered pharmacological response could result by a decrease in α1 subunit-containing GABAA receptors. Likewise, a decreased γ2 subunits support the formation of GABAA receptors in which this subunit might be substituted. Given that γ2 subunits are a necessary prerequisite for the formation of benzodiazepine-sensitive GABAA receptors (89; 90), the lack of anxiolytic activity of diazepam may result from the formation of benzodiazepine-insensitive GABAA receptors in neuronal circuits that regulate anxiety (39; 91).

Increases of 4 subunit-containing GABAA receptor expression in the frontal cortex appeared to be irrelevant to the behavioral or pharmacological alterations observed in socially isolated mice. GABA agonists such as THIP or the allosteric modulator, allopregnanolone, show selectivity and increased potency, respectively, for GABAA receptors containing 4/-subunits. These compounds comparably decrease locomotor activity in group-housed and socially isolated mice (91). In contrast to diazepam, allopregnanolone dose-dependently induces potent anxiolytic actions in socially isolated mice (16; 39).

Interestingly, the expression of GABAA receptor subunits is susceptible to changes in brain neurosteroid levels. In particular, expression of α4−containing subunits increases during progesterone withdrawal or following blockade of 5α−reductase (93). Likewise, in socially isolated mice, allopregnanolone levels decrease in several corticolimbic structures that concomitantly show changes in GABAA receptor subunit mRNA and protein expression. It would be important to determine whether social isolation directly affects the expression of GABAA receptor subunit composition or whether such changes are mediated by decreasing the levels of 5α−DHP and its binding at nuclear progesterone receptors or by allopregnanolone biosynthesis downregulation.

Advertisement

8. Selective brain steroidogenic stimulants (SBSSs) improve behavioral deficits in socially isolated mice

Behavioral deficits induced by social isolation in rodents include aggressive behavior (94-96). Aggression is correlated with the downregulation of corticolimbic allopregnanolone biosynthesis (35). Upregulation of allopregnanolone levels in socially isolated mice by systemic administration or local microinfusion of allopregnanolone induces a dose-dependent amelioration of aggressive behavior of a resident mouse to a same-sex intruder (35; 77). Thus, the decrease of corticolimbic allopregnanolone levels appears to be involved in the expression of aggression.

As indicated above, SSRI antidepressants potently increase the levels of allopregnanolone in rodents and depressed humans. The effects of paroxetine and fluoxetine on allopregnanolone levels were independent from pregnenolone or progesterone levels that failed to change (34; 76). Racemic fluoxetine (R- and S-isomers) normalized the righting reflex loss induced by pentobarbital in mice by increasing corticolimbic allopregnanolone levels (35-37). Of note, at the doses used, fluoxetine failed to change the behavior and allopregnanolone levels of group housed mice (35; 36). Importantly, inhibition of serotonin synthesis by treatment with p-chlorophenylalanine failed to block the behavioral effects of fluoxetine, suggesting that increasing corticolimbic allopregnanolone levels is part of the pharmacological actions of fluoxetine (76).

These observations led us to hypothesize that fluoxetine could improve the behavioral abnormalities of socially isolated mice by enhancing corticolimbic allopregnanolone biosynthesis rather than by inhibiting serotonin reuptake. This hypothesis was investigated using the R- and S-stereoisomers of fluoxetine and norfluoxetine as pharmacological tools. We expected that these drugs would stereospecifically upregulate corticolimbic allopregnanolone content but have no stereoselectivity with regard to inhibition of 5-HT reuptake. We additionally thought that doses of fluoxetine and norfluoxetine stereoisomers that increase corticolimbic allopregnanolone content might differ from those that inhibit 5-HT reuptake. Indeed (16; 35-38), fluoxetine dose-dependently and stereospecifically normalized the duration of pentobarbital-induced sedation and reduced aggressiveness, fear responses, and anxiety-like behavior at the same submicromolar doses that normalized the downregulation of brain allopregnanolone content in socially isolated mice. Interestingly, the S-stereoisomers of fluoxetine or norfluoxetine appeared to be 3 to 7 fold more potent than their respective R-stereoisomers and S-norfluoxetine was about 5-fold more potent than S-fluoxetine. Importantly, the effective concentrations (EC50s) of S-fluoxetine and S-norfluoxetine that normalize the brain allopregnanolone content are 10- (S-fluoxetine) and 50-fold (S-norfluoxetine) lower than their respective EC50s needed to inhibit 5-HT reuptake (35-38). Remarkably, the SSRI activity of S or R-fluoxetine and of S or R-norfluoxetine was devoid of stereospecificity (35; 36). Hence, this study demonstrated that neither the behavioral action nor the normalization of corticolimbic allopregnanolone content by S-fluoxetine and S-norfluoxetine is related to their intrinsic SSRI activity.

Advertisement

9. A novel promising therapy for anxiety disorders and PTSD

In the pathophysiology of depression and PTSD, a GABAergic neurotransmission dysfunction could at least in part be involved in the symptomatology of these disorders. Decreased GABA levels and reductions in GABAA and GABAB receptor binding and/or sensitivity have been found in depressed patients (97; 98). In PTSD, decreased frontal lobe benzodiazepine receptor binding (99; 100) and decreased plasma GABA levels (101) have been demonstrated. These changes were most consistently and profoundly observed among treatment resistant patients. Benzodiazepines have not been found to effectively treat PTSD (10-12) and SSRIs sertraline and paroxetine are the only medications currently approved by the Federal Drug Administration (FDA) for the treatment of PTSD. However, their effect sizes are modest (102-105), or even ineffective (106). In patients who cannot adequately synthesize allopregnanolone and in whom administration of an SSRI (or SBSS) is ineffective, the administration of an allopregnanolone analog (e.g. 107, 108), such as ganaxolone may offer a therapeutic alternative. A multisite Phase II trial of the efficacy and safety of ganaxolone in PTSD is currently been tested. Other medications that increase plasma allopregnanolone levels by a different mechanisms than the SSRIs also may be effective in PTSD (109-111).

The findings that the socially isolated mouse expresses decreased levels of allopregnanolone, as well as changes in the expression of several GABAA receptor subunits in corticolimbic structures that regulate cognition, anxiety, PTSD, and depression suggests that the socially isolated mouse model may be useful in investigating new molecules designed to improve behavioral deficits characterized by GABAA receptor signal transduction dysfunction (reviewed in 16; 38; 73).

Hence, as in PTSD patients, the socially isolated mouse fails to respond to sedative and anxiolytic benzodiazepines. Our studies demonstrate that allopregnanolone or S-norfluoxetine -at nonserotonergic doses- infused into the basolateral amygdala potently increase allopregnanolone biosynthesis in target corticolimbic areas including the hippocampus, basolateral amygdala, and frontal cortex (77) and exert a strong anti-anxiety, anti-fear, and anti-aggression effect (35-38; 72; 77).

Neurosteroids lack GABAA receptor subunit selectivity and the functional GABAA receptor binding characteristics of benzodiazepines. Thus, this suggests that allopregnanolone, its analogs, or molecules that stimulate allopregnanolone biosynthesis might be advantageous over benzodiazepines in a scenario of neurosteroid downregulation and changes in GABAA receptor subunit subtypes. Despite benzodiazepines, allopregnanolone activates GABAA receptors incorporating α4, α6, and δ subunits in combination with γ and β subunits (64-66). Thus, allopregnanolone or SBSSs improve anxiety, fear, and aggressiveness when benzodiazepines fail. Of note and in contrast to benzodiazepines, both allopregnanolone and SBSS molecules decrease anxiety, fear, and aggression at concentrations that fail to be sedative (16; 35; 39; 77).

New SBSS molecules that fail to exert any significant SSRI activity but increase corticolimbic allopregnanolone levels and thereby improve behavioral symptoms in mouse models of anxiety and depression. The high potency and stereospecificity of these drugs in reducing behavioral deficits and in normalizing brain allopregnanolone content suggest that they may affect specific targets for regulating neurosteroidogenesis. The finding that protracted social isolation affects the expression of 5α-reductase in corticolimbic structures, but fails to change the expression of 3α-HSD, as well as the finding that brain progesterone levels don’t change in socially isolated mice suggest that a mechanism involving 5α-reductase is responsible for the decrease of corticolimbic allopregnanolone content. This is further supported by the fact that 5α-reductase is the rate-limiting step-enzyme in allopregnanolone biosynthesis from progesterone (73). Hence, these data suggest that fluoxetine and norfluoxetine mediate upregulation of corticolimbic allopregnanolone levels by a direct action on 5α-reductase. However, in vitro studies by Griffin and Mellon (112) showed that fluoxetine, paroxetine, and sertraline failed to activate 5α-reductase and instead, directly activated 3α-HSD by decreasing its Km for 5α-DHP, thereby facilitating an accumulation of allopregnanolone (112). The hypothesis that neurosteroidogenic antidepressants activate 3α-HSD is also suggested by the finding that fluoxetine accelerates the rate of allopregnanolone accumulation during incubation of brain slices with 5α-DHP (34). Furthermore, progesterone levels in group-housed and socially isolated mice are not affected by fluoxetine administration, suggesting that the SSRI/SBSSs impact neurosteroidogenesis downstream from progesterone (34; 76). On the other hand, experiments by Trauger and collaborators (113) 1were inconsistent with the hypothesis that fluoxetine and paroxetine directly activate 3α-HSD. The finding that low doses of the S isomers of fluoxetine or norfluoxetine increase corticolimbic levels of allopregnanolone in socially isolated mice, but fail to change levels in group-housed mice, suggests that 5α-reductase and/or 3α-HSD may become more susceptible to the effects of SBSSs during isolation (reviewed in 38). Investigations at the molecular enzymatic level will clarify whether social isolation and neurosteroidogenic agents change the kinetics of 5α-reductase and/or 3α-HSD.

Other feasible pharmacological targets to enhance neurosteroidogenesis include the translocase protein (18 kDa) or TSPO, previously called mitochondrial peripheral benzodiazepine receptor or PBR (114). TSPO represents the starting point and an important rate-limiting step in neurosteroidogenesis. It gives access to neurosteroids in the brain by regulating the entry of cholesterol into the inner mitochondrial membranes and its conversion to pregnenolone by P450scc, which is located in the inner mitochondrial membrane (29; 114). A cascade of enzymatic processes then take place in the cytosol, resulting in the production of neuroactive steroids, including pregnenolone sulfate, DHEAS (though apparently not in human brain (115)], THDOC, and allopregnanolone (reviewed in 31).

New molecules that bind with high affinity to TSPO have been recently investigated. These drugs are able to exert important anxiolytic effects but are devoid of the unwanted side effects associated with benzodiazepines, including over-sedation and tolerance (28; 29). In mouse models, TSPO agents have been shown to potently increase pregnenolone levels in the hippocampus and cortex, as well as to induce anxiolytic effects (116-119). TSPO ligands include XBD173 and etifoxine, which have proven to be highly efficacious anxiolytic and antidepressant drugs in a number of behavioral tests (29; 30). The anxiolytic and antidepressant effects of these agents were related their ability to increase neurosteroid biosynthesis, as confirmed by studies in which key enzyme blockers for neurosteroid biosynthesis, including finansteride and trilostane (56; 30), were used. TSPO ligands have recently showed promising therapeutic effects in clinical studies (29; 30).

Advertisement

10. Closing remarks

The new class of drugs, the SBSSs (selective brain steroidogenic stimulants) -whose mechanism of action involves the stimulation of neurosteroidogenesis with the goal of increasing brain allopregnanolone levels- has emerged as a new therapeutic strategy for the treatment of psychiatric disorders associated with a downregulation of brain allopregnanolone biosynthesis. These disorders include anxiety, depression, and PTSD.

In comparison to benzodiazepines, the SBSSs are more efficacious as well as devoid of the unwanted side-effects induced by benzodiazepines. Allopregnanolone pharmacology involves the allosteric modulation of GABA action at GABAA receptors, which is broader than that of benzodiazepines, which fail to modulate GABAA receptors containing α4 and α6 subunits. Hence, selective stimulation of allopregnanolone biosynthesis may avoid the therapeutic hindrances caused by the formation of benzodiazepine-resistant GABAA receptors with altered subunit composition, such as may occur in stress-related psychiatric disorders (reviewed 6in 120). Thus, novel SBSS drugs that specifically increase corticolimbic allopregnanolone biosynthesis appear to be a novel promising pharmacological class of future drugs for the treatment of anxiety disorders, depression, and PTSD.

Acknowledgments

This review was supported by the National Institute of Mental Health Grant MH 085999 (to G. Pinna).

References

  1. 1. Diagnostic Statistical Manual of Mental Disorders.2000Washington DC: American Psychiatric Press;
  2. 2. BertonO.NestlerI. J.2006New approaches to antidepressant drug discovery: beyond monoamines. Nat Rev Neurosci ; 7137151
  3. 3. KesslerR. C.BerglundP.DemlerO.JinR.MerikangasK. R.WaltersE. E.2005Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry ; 62593602
  4. 4. CostaE.GuidottiA.1996Benzodiazepines on trial: a research strategy for their rehabilitation. Trends Pharmacol Sci ; 17192200
  5. 5. BandelowB.ZoharJ.HollanderE.KasperS.MöllerH. J.et al.2008World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders- first revision. World J Biol Psychiatry ; 9248312
  6. 6. BaldwinD. S.AndersonI. M.NuttD. J.BandelowB.BondA.DavidsonJ. R.denJ. A.BoerN. A.FinebergM.KnappJ.ScottH. U.Wittchen2005British Association for Psychopharmacology. Evidence-based guidelines for the pharmacological treatment of anxiety disorders: recommendations from the British Association for Psychopharmacology. J Psychopharmacol ; 1956796
  7. 7. RudolphU.MöhlerH.2006GABA-based therapeutic approaches: GABAA receptor subtype functions. Curr Opin Pharmacol; 61823
  8. 8. NuttD. J.BallengerJ. C.SheehanD.WittchenH. U.2002Generalized anxiety disorder: comorbidity, comparative biology and treatment. Int J Neuropsychopharmacol ; 531525
  9. 9. PinnaG.GaliciR.SchneiderH. H.StephensD. N.TurskiL.1997Alprazolam dependence prevented by substituting with the beta-carboline abecarnil. Proc Natl Acad Sci USA ; 94271923
  10. 10. ViolaJ.DitzlerT.BatzerW.HarazinJ.AdamsD.LettichL.BeriganT.1997Pharmacological management of post-traumatic stress disorder: clinical summary of a five-year retrospective study, Mil Med ; 1626169
  11. 11. DavidsonJ. R.2004Use of benzodiazepines in social anxiety disorder, generalized anxiety disorder, and posttraumatic stress disorder. J Clin Psychiatry ; 652933
  12. 12. GelpinE.BonneO.PeriT.BrandesD.ShalevA. Y.1996Treatment of recent trauma survivors with benzodiazepines: a prospective study. J Clin Psychiatry ; 573904
  13. 13. AtackJ. R.2003Anxioselective compounds acting at the GABA(A) receptor benzodiazepine binding site. Curr Drug Targets CNS Neurol Disord ; 221332
  14. 14. CostaE.1998From GABAA receptor diversity emerges a unified vision of GABAergic inhibition. Annu Rev Pharmacol Toxicol ; 3832150
  15. 15. PuiaG.1992Molecular mechanisms of the partial allosteric modulatory effects of bretazenil at gamma-aminobutyric acid type A receptor. Proc Natl Acad Sci USA ; 8936204
  16. 16. PinnaG.CostaE.GuidottiA.2006Fluoxetine and norfluoxetine stereospecifically and selectively increase brain neurosteroid content at doses that are inactive on 5-HT reuptake. Psychopharmacology ; 186362372
  17. 17. RasmussonA. M.PinnaG.PaliwalP.WeismanD.GottschalkC.CharneyD.et al.2006Decreased cerebrospinal fluid allopregnanolone levels in women with posttraumatic stress disorder. Biol Psychiatry ; 6070413
  18. 18. NappiR.E. PetragliaF.LuisiS. PolattiF. FarinaC.GenazzaniA.R. 2001. Serum allopregnanolone in women with postpartum “blues”. Obstet Gynecol ; 97: 77-80.
  19. 19. StröhleA.RomeoE. di MicheleF.PasiniA.YassouridisA. HolsboerF. RupprechtR. 2002. GABAA receptor-modulationg neuroactive steroid composition in patients with panic disorder before and during paroxetine treatment. Am J Psychiatry ; 159: 145-7.
  20. 20. RapkinA. J.MorganM.GoldmanL.BrannD. W.SimoneD.MaheshV. B.1997Progesterone metabolite allopregnanolone in women with presmenstrual syndrome. Obstet Gynecol ; 9070914
  21. 21. 34188590321. C.E. Marx, R.S. Keefe, R.W. Buchanan, R.M. Hamer, J.D. Kilts, D.W. Bradford, et al. 2009. Proof-of-concept trial with the neurosteroid pregnenolone targeting cognitive and negative symptoms in schizophrenia. Neuropsycopharmacology ; 34: 1885-903
  22. 22. PearlsteinT.2010Premenstrual dysphoric disorder: out of the appendix. Arch Womens Ment Health. ; 13213
  23. 23. AminZ.MasonG. F.CavusI.KrystalJ. H.RothmanD. L.EppersonC. N.2007The interaction of neuroactive steroids and GABA in the development of neuropsychiatric disorders in women. J Psychopharmacol ; 2141420
  24. 24. BäckströmT. AndreenL. BirznieceV. BjörnI. et al. 2003. The role of hormones and hormonal treatments in premenstrual syndrome. CNS Drugs. ; 17: 325-42.
  25. 25. BlochM.SchmidtP. J.DanaceauM.MurphyJ.NiemanL.RubinowD. R.2000Effects of gonadal steroids in women with a history of postpartum depression. Am J Psychiatry ; 15792430
  26. 26. UzunovaV.ShelineY.DavisJ. M.RasmussonA.UzunovD. P.CostaE.GuidottiA.1998Increase in the cerebrospinal fluid content of neurosteroids in patients with unipolar major depression who are receiving fluoxetine or fluvoxamine. Proc Natl Acad Sci USA ; 95323944
  27. 27. RomeoE.StröhleA.SpallettaG.di MicheleF.HermannB.HolsboerF.PasiniA.RupprechtR.1998Effects of antidepressant treatment on neuroactive steroids in major depression. Am J Psychiatry ; 1559103
  28. 28. RupprechtR.RammesG.EserD.BaghaiT.C. et al. 2009. Translocator protein (18 kDa) as target for anxiolytics without benzodiazepine-like side effects. Science. ; 325: 490-3.
  29. 29. RupprechtR.PapadopoulosV.RammesG.BaghaiT. C.FanJ.AkulaN.GroyerG.AdamsD.SchumacherM.2010Translocator protein (18 kDa) (TSPO) as a therapeutic target for neurological and psychiatric disorders. Nat Rev Drug Discov. ; 997188
  30. 30. SchüleC.EserD.BaghaiT. C.NothdurfterC.KesslerJ. S.RupprechtR.2011Neuroactive steroids in affective disorders: target for novel antidepressant or anxiolytic drugs? Neuroscience Mar 23. [Epub ahead of print]
  31. 31. CostaB.DaE.PozzoB.ChelliN.SimolaM.MorelliM.LuisiM.MaccheroniS.TalianiF.SimoriniF.DaSettimo. C.Martini2011Anxiolytic properties of a 2phenylindolglyoxylamide TSPO ligand: Stimulation of in vitro neurosteroid production affecting GABAA receptor activity. Psychoneuroendocrinology ; 36; 463-472.
  32. 32. Agis-BalboaR. C.GuidottiA.WhitfieldH.PinnaG.2010Allopregnanolone biosynthesis is downregulated in the prefrontal cortex/ Brodmann’s area 9 (BA9) of depressed patients. Neuroscience Meeting Planner. San Diego, CA: Society for Neuroscience,
  33. 33. van BroekhovenF.VerkesR. J.2003Neurosteroids in depression: a review. Psychopharmacology ; 16597110
  34. 34. UzunovD. P.CooperT. B.CostaE.GuidottiA.1996Fluoxetine elicited changes in brain neurosteroid content measured by negative ion mass fragmentography. Proc Natl Acad Sci USA ; 931259912604
  35. 35. PinnaG.DongE. MatsumotoK. CostaE. GuidottiA. 2003. In socially isolated mice, the reversal of brain allopregnanolone down-regulation mediates the anti-aggressive action of fluoxetine. Proc Natl Acad Sci USA ; 10020352040
  36. 36. PinnaG.CostaE.GuidottiA.2004Fluoxetine and norfluoxetine stereospecifically facilitate pentobarbital sedation by increasing neurosteroids. Proc Natl Acad Sci USA ; 10162226225
  37. 37. PinnaG.CostaE.GuidottiA.2009SSRIs act as selective brain steroidogenic stimulants (SBSSs) at low doses that are inactive on 5-HT reuptake. Curr Opin Pharmacol ; 92430
  38. 38. PinnaG.2010In a mouse model relevant for post-traumatic stress disorder, selective brain steroidogenic stimulants (SBSS) improve behavioral deficits by normalizing allopregnanolone biosynthesis. Behav Pharmacol. ; 2143850
  39. 39. NinM.SchulerL. A.MartinezR.ThomasM.NelsonG.Pinna2011Allopregnanolone and S-norfluoxetine decrease anxiety-like behavior in a mouse model of anxiety/depression. Trabajos del Instituto Cajal. ; 83215216
  40. 40. BaulieuE. E.1981Steroid hormones in the brain: several mechanisms. In: Steroid hormone regulation of the brain. Fuxe K, Gustafson JA, Wettenberg L (editors). ; Elmsford, NY: Pergamon; 314
  41. 41. BaulieuE. E.RobelP.SchumacherM.2001Neurosteroids: beginning of the story. Int Rev Neurobiol ; 46132
  42. 42. CheneyD. L.UzunovD.CostaE.GuidottiA.1995Gas chromatographic-mass fragmentographic quantitation of 3α-hydroxy-5α-pregnan-20-one (allopregnanolone) and its precursors in blood and brain of adrenalectomized and castrated rats. J Neurosci ; 1546414650
  43. 43. GuidottiA.DongE.MatsumotoK.PinnaG.RasmussonA. M.CostaE.2001The socially-isolated mouse: a model to study the putative role of allopregnanolone and 5α-dihydroprogesterone in psychiatric disorders. Brain Res Rev ; 37110115
  44. 44. Stoffel-WagnerB.2001Neurosteroid metabolism in the human brain. European Journal of Endocrinology ; 145669679
  45. 45. PuiaG.SantiM. R.ViciniS.PritchettD. B.PurdyR. H.PaulS. M.SeeburgP. H.CostaE.1990Neurosteroids act on recombinant human GABAA receptors. Neuron ; 4759765
  46. 46. PuiaG.ViciniS.SeeburgP. H.CostaE.1991Influence of recombinant gamma-aminobutyric acid-a receptor subunit composition on the action of allosteric modulators of gammaaminobutyricacid-gated Cl-currents. Mol Pharmacol ; 39691696
  47. 47. PuiaG.MienvilleJ. M.MatsumotoK.TakahataH.WatanabeH.CostaE.GuidottiA.2003On the putative physiological role of allopregnanolone on GABAA receptor function. Neuropharmacology 444955
  48. 48. PinnaG.UzunovaV.MatsumotoK.PuiaG.MienvilleJ. M.CostaE.GuidottiA.2000Brain allopregnanolone regulates the potency of the GABAA receptor agonist muscimol. Neuropharmacology ; 39440448
  49. 49. LambertJ. J.BelelliD.PedenD. R.VardyA. W.PetersJ. A.2003Neurosteroid modulation of GABAA receptors. Prog Neurobiol ; 716780
  50. 50. LambertJ. J.CooperM. A.SimmonsR. D.WeirC. J.BelelliD.2009Neurosteroids: endogenous allosteric modulators of GABAA receptors. Psychoneuroendocrinology ; 34 Suppl 1: S4858
  51. 51. BelelliD.LambertJ. J.2005Neurosteroids: endogenous regulators of the GABAA receptor. Nat Rev Neurosci ; 6565575
  52. 52. MajewskaM. D.1992Neurosteroids: endogenous bimodal modulators of the GABAA receptor. Mechanism of action and physiological significance. Prog Neurobiol ; 38379395
  53. 53. D’AquilaP. S.CanuS.SardellaM.SpanuC.SerraG.FranconiF.2010Dopamine is involved in the antidepressant-like effect of allopregnanolone in the forced swimming test in female rats. Behav Pharmacol ; 21218
  54. 54. NinM. S.SallesF. B.AzeredoL. A.FrazonA. P.GomezR.BarrosH. M.2008Antidepressant effect and changes of GABAA receptor gamma2 subunit mRNA after hippocampal administration of allopregnanolone in rats. J Psychopharmacol ; 2247785
  55. 55. Rodríguez-LandaJ. F.ContrerasC. M.García-RíosR. I.2009Allopregnanolone microinjected into the lateral septum or dorsal hippocampus reduces immobility in the forced swim test: participation of the GABAA receptor. Behav Pharmacol ; 20614622
  56. 56. Kita and Furukawa.2008Involvement of neurosteroids in the anxiolytic-like effects of AC-5216 in mice. Pharmacol Biochem Behav ; 891718
  57. 57. Lonsdale and Burnham.2007The anticonvulsant effects of allopregnanolone against amygdala-kindled seizures in female rats. Neurosci Lett ; 41114751
  58. 58. MaresP.MikuleckáA.HaugvicováR.KasalA.2006Anticonvulsant action of allopregnanolone in immature rats. Epilepsy Res ; 701107
  59. 59. Martin-GarciaE.PallaresM.2005The intrahippocampal administration of the neurosteroid allopregnanolone blocks the audiogenic seizures induced by nicotine. Brain Res ; 106214450
  60. 60. JainN. S.HiraniK.ChopdeC. T.2005Reversal of caffeine-induced anxiety by neurosteroid 3-alpha-hydroxy-5-alpha-pregnane-20-one in rats. Neuropharmacology ; 4862738
  61. 61. PuiaG.ViciniS.SeeburgP. H.CostaE.1992Different sites of action of neurosteroids and benzodiazepines on natural and recombinant GABAA receptors. Adv Biochem Psychopharmacol. ; 4710310
  62. 62. PuiaG.DucicI.ViciniS.CostaE.1993Does neurosteroid modulatory efficacy depend on GABAA receptor subunit composition? Receptors Channels. ; 113542
  63. 63. MatsumotoK.NomuraH.MurakamiY.TakiK.TakahataH.WatanabeH.2003Long-term social isolation enhances picrotoxin seizure susceptibility in mice: up-regulatory role of endogenous brain allopregnanolone in GABAergic systems. Pharm Biochem Behav 75831835
  64. 64. HosieA. M.WilkinsM. E.daH. M.SilvaT. G.Smart2006Endogenous neurosteroids regulate GABAA receptors through two discrete transmembrane sites. Nature ; 4444869
  65. 65. BelelliD.CasulaA.LingA.LambertJ. J.2002The influence of subunit composition on the interaction of neurosteroids with GABAA receptors. Neuropharmacology ; 4365161
  66. 66. ViciniS.1991Pharmacologic significance of the structural heterogeneity of the GABAA receptor-chloride ion channel complex. Neuropsychopharmacology ; 4915
  67. 67. Agis-BalboaR. C.PinnaG.ZhubiA.MalokuE.VeldicM.CostaE.GuidottiA.2006Characterization of brain neurons that express enzymes mediating neurosteroid biosynthesis. Proc Natl Acad Sci USA 1031460214607
  68. 68. AkkG.ShuH. J.WangC.SteinbachJ. H.ZorumskiC. F.CoveyD. F.MennerickS.2005Neurosteroid access to the GABAA receptor. J Neurosc ; 251160511613
  69. 69. PinaultD.2004The thalamic reticular nucleus: structure, function and concept. Brain Res Rev ; 46131
  70. 70. SerraM.PisuM. G.LitteraM.PapiG.SannaE.TuveriF.UsalaL.PurdyR. H.BiggioG.2000Social isolation-induced decreases in both the abundance of neuroactive steroids and GABA(A) receptor function in rat brain. J Neurochem. ; 7573240
  71. 71. BortolatoM.DevotoP.RoncadaP.FrauR.FloreG.SabaP.PistrittoG.SoggiuA.PisanuS.ZappalaA.RistaldiM. S.TattoliM.CuomoV.MarrosuF.BarbacciaM. L.2011Isolation rearing-induced reduction of brain 5α-reductase expression: Relevance to dopaminergic impairments. Neuropharmacology 60(7-8):1301-8
  72. 72. MatsumotoK.PuiaG.DongE.PinnaG. 2007. GABAA receptor neurotransmission dysfunction in a mouse model of social isolation-induced stress: possible insights into a non-serotonergic mechanism of action of SSRIs in mood and anxiety disorders. Stress ; 10: 3-12.
  73. 73. DongE.MatsumotoK.UzunovaV.SugayaI.CostaE.GuidottiA.2001Brain 5α-dihydroprogesterone and allopregnanolone synthesis in a mouse model of protracted social isolation. Proc Natl Acad Sci USA ; 9828492854
  74. 74. PibiriF.NelsonM.GuidottiA.CostaE.PinnaG.2008Decreased allopregnanolone content during social isolation enhances contextual fear: a model relevant for posttraumatic stress disorder. Proc Natl Acad Sci USA ; 10555675572
  75. 75. Agis-BalboaR. C.PinnaG.KadriuB.CostaE.GuidottiA.2007Downregulation of 5α-reductase type I mRNA expression in cortico-limbic glutamatergic circuits of mice socially isolated for four weeks. Proc Natl Acad Sci USA ; 1041873641
  76. 76. MatsumotoK.UzunovaV.PinnaG.TakiK.UzunovD. P.WatanabeH.MienvielleJ. M.GuidottiA.CostaE.1999Permissive role of brain allopregnanolone content in the regulation of pentobarbital-induced righting reflex loss. Neuropharmacology ; 38955963
  77. 77. NelsonM.PinnaG.2011S-norfluoxetine infused into the basolateral amygdala increases allopregnanolone levels and reduces aggression in socially isolated mice. Neuropharmacology ; 6011541159
  78. 78. RodgersR. J.JohnsonN. J.1998Behaviorally selective effects of neuroactive steroids on plus-maze anxiety in mice. Pharmacol Biochem Behav ; 5922132
  79. 79. FryeC. A.RhodeM. E.2006Infusions of 5alpha-pregnan-3alpha-ol-20-one (3alpha,5alpha-THP) to the ventral tegmental area, but not the substantia nigra, enhance exploratory, anti-anxiety, social and sexual behaviours and concomitantly increase 3alpha,5alpha-THP concentrations in the hippocampus, diencephalon and cortex of ovariectomised oestrogen-primed rats. J Neuroendocrinol ; 1896075
  80. 80. BitranD.HilversR. J.KelloggC. K.1991Anxiolytic effects of 3 alpha-hydroxy-5 alpha[beta]-pregnan-20-one: endogenous metabolites of progesterone that are active at the GABAA receptor. Brain Res ; 56115761
  81. 81. WielandS.LanN. C.MirasedeghiS.GeeK. W.1991Anxiolytic activity of the progesterone metabolite 5 alpha-pregnan-3 alpha-o1-20-one. Brain Res ; 5652638
  82. 82. DeoG. S.DandekarM. P.UpadhyaM. A.KokareD. M.SubhedarN. K.2010Neuropeptide Y Y1 receptors in the central nucleus of amygdala mediate the anxiolytic-like effect of allopregnanolone in mice: Behavioral and immunocytochemical evidences. Brain Res ; 13187786
  83. 83. EnginE.TreitD.2007The anxiolytic-like effects of allopregnanolone vary as a function of intracerebral microinfusion site: the amygdala, medial prefrontal cortex, or hippocampus. Behav Pharmacol ; 1846170
  84. 84. FryeC. A.ParisJ. J.RhodesM. E.2009Increasing 3alpha,5alpha-THP following inhibition of neurosteroid biosynthesis in the ventral tegmental area reinstates anti-anxiety, social, and sexual behavior of naturally receptive rats. Reproduction ; 13711928
  85. 85. AkbarianS.HuntsmanM. M.KimJ. J.TafazzoliA.PotkinS. G.BunneyW. E. J.JonesE. G.1995GABAA receptor subunit gene expression in human prefrontal cortex: comparison of schizophrenics and controls. Cereb Cortex 555060
  86. 86. DeanB.HussainT.HayesW.ScarrE.KitsoulisS.HillC.OpeskinK.CopolovD. L.1999Changes in serotonin2A and GABA(A) receptors in schizophrenia: studies on the human dorsolateral prefrontal cortex. J Neurochem ; 7215939
  87. 87. LewisD. A.2000GABAergic local circuit neurons and prefrontal cortical dysfunction in schizophrenia. Brain Res Brain Res Rev. ; 312706
  88. 88. IshikawaM. MizukamiK. IwakiriM.HidakaS.AsadaT.2004. GABAA receptor gamma subunits in the prefrontal cortex of patients with schizophrenia and bipolar disorder. Neuroreport ; 15180912
  89. 89. RudolphU.MöhlerH.2004Analysis of GABAA receptor function and dissection of the pharmacology of benzodiazepines and general anesthetics through mouse genetics. Annu Rev Pharmacol Toxicol. ; 4447598
  90. 90. RudolphU.CrestaniF.BenkeD.BrünigI.BensonJ. A.FritschyJ. M.MartinJ. R.BluethmannH.MöhlerH.1999Benzodiazepine actions mediated by specific gamma-aminobutyric acid(A) receptor subtypes. Nature ; 401796800
  91. 91. PinnaG.Agis-BalboaR. C.ZhubiA.MatsumotoK.GraysonD. R.CostaE.GuidottiA.2006Imidazenil and diazepam increase locomotor activity in mice exposed to protracted social isolation. Proc Natl Acad Sci USA ; 10342754280
  92. 92. CrestaniF. MartinJ.R. MöhlerH. RudolphU. 2000. Mechanism of action of the hypnotic zolpidem in vivo. Br J Pharmacol. ; 131: 1251-4.
  93. 93. SmithS. S.GongQ. H.HsuF. C.MarkowitzR. S.ffrench-MullenJ. M.LiX.1998GABA(A) receptor alpha4 subunit suppression prevents withdrawal properties of an endogenous steroid. Nature. ;392667992630
  94. 94. ValzelliL.1973The "isolation syndrome" in mice. Psychopharmacologia. ; 3130520
  95. 95. ValzelliL.1981Psychopharmacology of aggression: an overview. Int Pharmacopsychiatry ; 163948
  96. 96. MatsumotoK.CaiB.SatohT.OhtaH.WatanabeH.1991Desipramine enhances isolation-induced aggressive behavior in mice. Pharmacol Biochem Behav ; 3916770
  97. 97. SanacoraG.SaricicekA.2007GABAergic contributions to the pathophysiology of depression and the mechanism of antidepressant action. CNS Neurol Disord Drug Targets 6127140
  98. 98. SanacoraG.2010Cortical Inhibition, gamma-aminobutyric acid, and major depression: there is plenty of smoke but is there fire? Biological Psychiatry. ; 673978
  99. 99. BremnerJ. D.InnisR. B.SouthwickS. M.StaibL.ZoghbiS.CharneyD. S.2000Decreased benzodiazepine receptor binding in prefrontal cortex in combat-related posttraumatic stress disorder. Am J Psychiatry ; 15711206
  100. 100. FujitaM.SouthwickS. M.DenucciC. C.ZoghbiS. S.DillonM. S.BaldwinR. M.BozkurtA.KugayaA.VerhoeffN. P.SeibylJ. P.2004Innis RB: Central type benzodiazepine receptors in Gulf War veterans with posttraumatic stress disorder. Biol Psychiatry ; 5695100
  101. 101. VaivaG.ThomasP.DucrocqF.FontaineM.BossV.DevosP.RascleC.CottencinO.BrunetA.LaffargueP.GoudemandM.2004Low posttrauma GABA plasma levels as a predictive factor in the development of acute posttraumatic stress disorder. Biol Psychiatry ; 55:250-4,
  102. 102. BradyK. PearlsteinT. AsnisG.M. BakerD.RothbaumB. SikesC. FarfelG.M. 2000. Efficacy and safety of sertraline treatment of posttraumatic stress disorder. JAMA ; 28318371844
  103. 103. DavidsonJ. R. T.RothbaumB. O.van der KolkB. A.SikesC. R.FarfelG. M.2001Multicenter, double-blind comparison of sertraline and placebo in the treatment of posttraumatic stress disorder. Arch Gen Psychiatry ; 58485492
  104. 104. 104. R.D. Marshall, K.L. Beebe, M. Oldham, R. Zaninelli. 2001. Efficacy and safety of paroxetinetreatment for PTSD: A fixed-dose, placebo-controlled study. Am J Psychiatry ; 158:1982-1988
  105. 105. TuckerP.ZaninelliR.YehudaR.RuggieroL.DillinghamK.PittsC. D.2001Paroxetine in the treatment of chronic posttraumatic stress disorder: Results of a placebo-controlled, flexible-dosage study. J Clin Psychiatry ; 62860868
  106. 106. FriedmanM. J.MarmarC. R.BakerD. G.SikesC. R.FarfelG. M.2007Randomized, double-blind comparison of sertraline and placebo for posttraumatic stress disorder in a Department of Veterans Affairs setting. J Clin Psychiatry ; 68711720
  107. 107. GulinelloM.GongQ. H.SmithS. S.2003Progesterone withdrawal increases the anxiolytic actions of gaboxadol: role of alpha4betadelta GABA(A) receptors. Neuroreport ; 144346
  108. 108. KaminskiR. M.LivingoodM. R.RogawskiM. A.2004Allopregnanolone analogs that positively modulate GABA receptors protect against partial seizures induced by 6-Hz electrical stimulation in mice. Epilepsia ; 458647
  109. 109. MarxC. E.Van DorenM. J.DuncanG. E.LiebermanJ. A.MorrowA. L.2003Olanzapine and clozapine increase the GABAergic neuroactive steroid allopregnanolone in rodents. Neuropsychopharm ; 28113
  110. 110. GenazzaniA.D. Stomati M. BernardiF. PieriM. RovatiL. GenazzaniA.R. 2003. Long-term low-dose dehydroepiandrosterone oral supplementation in early and late postmenopausal women modulates endocrine parameters and synthesis of neuroactive steroids. Fertility & Sterility ; 80: 1495-501.
  111. 111. Schmidt P.J. DalyR.C. BlochM.SmithM.J. DanaceauM.A. SimpsonL.S.C. MurphyJ.H. HaqN. RubinowD.R. 2005. Dehydroepiandrosterone monotherapy in midlife-onset major and minor depression. Arch Gen Psychiatry ; 62: 154-162.
  112. 112. GriffinL. D.MellonS. H.1999Selective serotonin reuptake inhibitors directly alter activity of neurosteroidogenic enzymes. Proc Natl Acad Sci USA ; 96135127
  113. 113. TraugerJ. W.JiangA.StearnsB. A.Lo. P. V.Grasso2002Kinetics of allopregnanolone formation catalyzed by human 3 alpha hydroxysteroid dehydrogenase Type III (AKR1C2). Biochemistry ; 411345113459
  114. 114. PapadopoulosV.BaraldiM.GuilarteT. R.KnudsenT. B.LacapèreJ. J.LindemannP.NorenbergM. D.NuttD.WeizmanA.ZhangM. R.GavishM.2006Translocator protein (18kDa): new nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol Sci ; 274029
  115. 115. CompagnoneN. A.MellonS. H.2000Neurosteroids: biosynthesis and function of these novel neuromodulators. Front Neuroendocrinol ; 21156
  116. 116. ReddyD. S.KulkarniS. K.1996Role of GABA-A and mitochondrial diazepam binding inhibitor receptors in the anti-stress activity of neurosteroids in mice. Psychopharmacology ; 12828092
  117. 117. RomeoE.AutaJ.KozikowskiA. P.MaPapadopoulosD. V.PuiaG.CostaE.GuidottiA.1992Aryl-3-indoleacetamides (FGIN-1): a new class of potent and specific ligands for the mitochondrial DBI receptor (MDR). J Pharmacol Exp Ther ; 2629718
  118. 118. KorneyevA.PanB. S.PoloA.RomeoE.GuidottiA.CostaE.1993Stimulation of brain pregnenolone synthesis by mitochondrial diazepam binding inhibitor receptor ligands in vivo. J Neurochem ; 61151524
  119. 119. KitaA.KohayakawaH.KinoshitaT.OchiY.NakamichiK.KurumiyaS.FurukawaK.OkaM.2004Antianxiety and antidepressant-like effects of AC-5216, a novel mitochondrial benzodiazepine receptor ligand. Br J Pharmacol. ; 142105972
  120. 120. MaguireJ.ModyI.2009Steroid hormone fluctuations and GABAA R plasticity. Psychoneuroendocrinology ; 34: Suppl 1: S8490

Written By

Graziano Pinna

Submitted: 27 October 2010 Published: 01 August 2011