Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 97
1.
Article Ru | MEDLINE | ID: mdl-37084362

Despite the proven importance of neurosteroids in many physiological processes, their role in the pathogenesis of the most of psychiatric disorders remains relatively understudied. This article reviews the current clinical evidence on the effects of neurosteroids on the formation and treatment of anxiety disorder, depression, bipolar disorder, and schizophrenia. In particular, the article points out the ambivalent nature of the effects of neurosteroids on GABAA- and other receptors. We are especially interested in the anxiolytic and anxiogenic effects of some neurosteroids, the antidepressant effect of allopregnanolone in treating postpartum and other forms of depression, and the nature of short- and long-term mechanisms of antidepressant effects of neurosteroids of different types. The currently unproven hypothesis about the effect of changes in the level of neurosteroids on the course of bipolar disorder is also discussed, with an analysis of the scientific evidence on the development of schizophrenic symptomatology in relation to changing neurosteroid levels in the context of positive and cognitive symptoms.


Bipolar Disorder , Mental Disorders , Neurosteroids , Female , Humans , Neurosteroids/therapeutic use , Anxiety Disorders , Pregnanolone/therapeutic use , Pregnanolone/pharmacology , Pregnanolone/physiology
2.
Nat Neurosci ; 24(10): 1392-1401, 2021 10.
Article En | MEDLINE | ID: mdl-34400844

Compromised placental function or premature loss has been linked to diverse neurodevelopmental disorders. Here we show that placenta allopregnanolone (ALLO), a progesterone-derived GABA-A receptor (GABAAR) modulator, reduction alters neurodevelopment in a sex-linked manner. A new conditional mouse model, in which the gene encoding ALLO's synthetic enzyme (akr1c14) is specifically deleted in trophoblasts, directly demonstrated that placental ALLO insufficiency led to cerebellar white matter abnormalities that correlated with autistic-like behavior only in male offspring. A single injection of ALLO or muscimol, a GABAAR agonist, during late gestation abolished these alterations. Comparison of male and female human preterm infant cerebellum also showed sex-linked myelination marker alteration, suggesting similarities between mouse placental ALLO insufficiency and human preterm brain development. This study reveals a new role for a placental hormone in shaping brain regions and behaviors in a sex-linked manner. Placental hormone replacement might offer novel therapeutic opportunities to prevent later neurobehavioral disorders.


Cerebellum/growth & development , Endocrine Glands/physiology , Placenta/physiology , Pregnanolone/deficiency , Pregnanolone/physiology , Social Behavior , Aldehyde Reductase/genetics , Animals , Autism Spectrum Disorder/etiology , Cerebellum/physiology , Female , GABA Agonists/pharmacology , GABA Modulators , Gene Deletion , Humans , Infant , Infant, Newborn , Male , Mice , Muscimol/pharmacology , Pregnancy , Receptors, GABA-A/physiology , Sex Characteristics , Trophoblasts/metabolism , White Matter/pathology
3.
Int J Mol Sci ; 22(12)2021 Jun 19.
Article En | MEDLINE | ID: mdl-34205405

Oxygen causes white matter damage in preterm infants and male sex is a major risk factor for poor neurological outcome, which speculates the role of steroid hormones in sex-based differences. Preterm birth is accompanied by a drop in 17ß-estradiol (E2) and progesterone along with increased levels of fetal zone steroids (FZS). We performed a sex-based analysis on the FZS concentration differences in urine samples collected from preterm and term infants. We show that, in preterm urine samples, the total concentration of FZS, and in particular the 16α-OH-DHEA concentration, is significantly higher in ill female infants as compared to males. Since we previously identified Nup133 as a novel target protein affected by hyperoxia, here we studied the effect of FZS, allopregnanolone (Allo) and E2 on differentiation and Nup133 signaling using mouse-derived primary oligodendrocyte progenitor cells (OPCs). We show that the steroids could reverse the effect of hyperoxia-mediated downregulation of Nup133 in cultured male OPCs. The addition of FZS and E2 protected cells from oxidative stress. However, E2, in presence of 16α-OH-DHEA, showed a negative effect on male cells. These results assert the importance of sex-based differences and their potential implications in preterm stress response.


Dehydroepiandrosterone/analogs & derivatives , Estradiol/physiology , Infant, Premature/metabolism , Oligodendrocyte Precursor Cells/physiology , Pregnanolone/physiology , Sex Characteristics , Animals , Dehydroepiandrosterone/urine , Female , Humans , Infant, Newborn , Male , Mice , Minor Histocompatibility Antigens/metabolism , Nuclear Pore Complex Proteins/metabolism , Oxidative Stress
4.
Int J Mol Sci ; 22(11)2021 May 23.
Article En | MEDLINE | ID: mdl-34071053

Under stressful conditions, the hypothalamic-pituitary-adrenal (HPA) axis acts to promote transitory physiological adaptations that are often resolved after the stressful stimulus is no longer present. In addition to corticosteroids (e.g., cortisol), the neurosteroid allopregnanolone (3α,5α-tetrahydroprogesterone, 3α-hydroxy-5α-pregnan-20-one) participates in negative feedback mechanisms that restore homeostasis. Chronic, repeated exposure to stress impairs the responsivity of the HPA axis and dampens allopregnanolone levels, participating in the etiopathology of psychiatric disorders, such as major depressive disorder (MDD) and post-traumatic stress disorder (PTSD). MDD and PTSD patients present abnormalities in the HPA axis regulation, such as altered cortisol levels or failure to suppress cortisol release in the dexamethasone suppression test. Herein, we review the neurophysiological role of allopregnanolone both as a potent and positive GABAergic neuromodulator but also in its capacity of inhibiting the HPA axis. The allopregnanolone function in the mechanisms that recapitulate stress-induced pathophysiology, including MDD and PTSD, and its potential as both a treatment target and as a biomarker for these disorders is discussed.


Depressive Disorder, Major/physiopathology , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Pregnanolone/physiology , Adaptation, Physiological , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Chronic Disease , Corticosterone/metabolism , Depressive Disorder, Major/drug therapy , Feedback, Physiological , Female , GABA-A Receptor Agonists/therapeutic use , Humans , Male , Models, Biological , Pregnanolone/biosynthesis , Receptors, GABA-A/physiology , Sex Characteristics , Stress Disorders, Post-Traumatic/physiopathology , Stress, Physiological , Stress, Psychological/physiopathology , Stress, Psychological/psychology , gamma-Aminobutyric Acid/physiology
5.
Endocrinol Diabetes Metab ; 4(2): e00216, 2021 04.
Article En | MEDLINE | ID: mdl-33855217

Introduction: The neurosteroid allopregnanolone modulates oxytocin expression in the brain, and its effects arise from its action on the GABAA receptor. Whether neurosteroid levels and the function of the GABAA receptor are involved in the risk of preterm labour in pregnant women is unknown. Methods: Pregnant women with (n = 16) or without (n = 20) threatened preterm labour (TPL) in gestational week 33 + 6 days to 37 + 0 days were studied prospectively with procedures including foetal heart rate monitoring, vaginal examination, ultrasound examination and blood tests to determine allopregnanolone, progesterone and oxytocin levels. The GABAA receptor function in both groups was measured with a saccadic eye velocity test (SEVT). Results: Plasma oxytocin levels were higher in the TPL group than in the control group (41.5 vs. 37.0 pmol/L, respectively, p = .021). Although the allopregnanolone and progesterone levels in both groups did not differ, there was a negative association between blood oxytocin and allopregnanolone (as predictor) levels in the TPL group (B: -3.2, 95% confidence interval (CI): -5.5 to -0.9, p = .012). As a predictor of TPL, progesterone was associated with cervix maturity (odds ratio: 1.02, 95% CI: 1.00-1.04, p = .038). SEVT showed that the women in both groups had similar GABAA receptor functions. In both groups, body mass index correlated with peak saccadic eye velocity (r = .34, p = .044) and negatively with allopregnanolone (r = -.41, p = .013). Conclusions: Neurosteroid levels were unchanged in the peripheral blood of women with TPL, despite the increase in available oxytocin. Although the function of the GABAA receptor was unchanged in women with TPL, to ensure reliable results, saccadic eye velocity should be investigated during a challenge test with a GABAA receptor agonist.


Obstetric Labor, Premature/etiology , Pregnanolone/physiology , Adolescent , Adult , Biomarkers , Body Mass Index , Brain/metabolism , Female , Humans , Male , Obstetric Labor, Premature/diagnosis , Obstetric Labor, Premature/prevention & control , Oxytocin/blood , Oxytocin/metabolism , Pregnancy , Pregnanolone/blood , Progesterone/blood , Progesterone/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-A/physiology , Risk , Saccades , Young Adult
6.
Neuropeptides ; 76: 101937, 2019 Aug.
Article En | MEDLINE | ID: mdl-31253440

The progesterone metabolite, allopregnanolone (AlloP), is a GABAA receptor modulating steroid and is known to have orexigenic and pro-obesity effects. The neurobiological mechanisms underpinning these effects are most likely due to enhanced GABAergic signaling in the lateral arcuate nucleus (ARC) and medial paraventricular nucleus (PVN) of the hypothalamus. Inspired by the finding that GABAergic signaling is also important for the orexigenic effects of the circulating hormone, ghrelin, we sought to determine the extent to which AlloP (one of the most potent endogenous GABAA-receptor modulators) operates alongside ghrelin to enhance food intake. Male rats with ad libitum access to standard chow were injected intravenously with AlloP and/or ghrelin, alone or in combination. The intake of the standard chow was greater after AlloP 1 mg/kg together with ghrelin 30 µg/kg than with 30 µg/kg ghrelin alone. Food intake was also increased for the combined treatment of AlloP 0.5 mg/kg + ghrelin 10 µg/kg, AlloP 1 mg/kg + ghrelin 10 µg/kg, and AlloP 0.5 mg/kg + ghrelin 30 µg/kg. There was no significant difference in food intake between the two ghrelin doses or between the two doses of AlloP and the vehicle. In electrophysiological studies, physiologically relevant concentrations of AlloP prolonged the current decay time of spontaneous inhibitory post-synaptic current of dissociated cells of the ARC and PVN. We conclude that AlloP enhances the hyperphagic effect of ghrelin, findings of potential relevance for the hyperphagia associated with the luteal phase of the reproductive cycle.


Arcuate Nucleus of Hypothalamus/drug effects , Eating/drug effects , Ghrelin/administration & dosage , Paraventricular Hypothalamic Nucleus/drug effects , Pregnanolone/administration & dosage , Animals , Arcuate Nucleus of Hypothalamus/physiology , Ghrelin/physiology , Inhibitory Postsynaptic Potentials/drug effects , Male , Paraventricular Hypothalamic Nucleus/physiology , Pregnanolone/physiology , Rats, Wistar
7.
J Pharmacol Sci ; 139(2): 77-83, 2019 Feb.
Article En | MEDLINE | ID: mdl-30579760

The beneficial effects of physical activity for pain are denominated exercise-induced hypoalgesia (EIH). Here, we examined the age-related change and potential role of the neurosteroid allopregnanolone (ALLO) on EIH in rats. Adult and aged rats were randomly divided into one of three groups; non-exercise control, Low-exercise, and High-exercise. The animals in the Low- and High-exercise groups were subjected to a 10-minute treadmill workout at 40% and 80% maximum oxygen intake intensity, respectively. In the Low-exercise groups, a significant EIH response was observed in aged but not in adult rats. The pre-treatment with ALLO synthesis inhibitor finasteride, but not opioid-receptor antagonist naloxone, inhibited the Low-exercise induced EIH response in aged rats. Furthermore, the Low-exercise increased brain ALLO levels in aged animals compared with controls, which was correlated with the mechanical pain sensitivity. On the other hand, High-exercise could induce EIH response in both adult and aged animals, but it was more effective in adult rats. The pre-treatment with naloxone, but not finasteride, reduced the EIH observed after High-exercise in both adult and aged rats. Our findings demonstrated that effective EIH can be achieved even by mild-intensity exercise in aged animals via an increase of the brain ALLO levels.


Aging/physiology , Pain/physiopathology , Physical Conditioning, Animal/physiology , Pregnanolone/physiology , Animals , Behavior, Animal , Brain/metabolism , Male , Pain Threshold , Progesterone/metabolism , Rats, Wistar , Reflex , Spinal Cord/metabolism
8.
J Matern Fetal Neonatal Med ; 31(9): 1241-1245, 2018 May.
Article En | MEDLINE | ID: mdl-28337942

Fetal pain and fetal anesthesia are still matter of debate: some authors hypothesize that several intrauterine endocrine neuroinhibitors (ENIn) anesthetize the fetus, keeping it in a constant state of sleep, and making pharmacological fetal anesthesia useless for fetal surgery, while others argue fetal pain is possible and shoud be prevented with fetal anesthesy. AIM: To retrieve evidences about fetal pain, fetal arousability and about the level of sedation induced by the ENIn, in order to assess the necessity of direct fetal anesthesia during prenatal fetal surgery. METHODS: We performed a careful literature review (1990-2016) on fetal arousability, and on the possibility that ENIn at the average fetal blood levels induce actual anesthesia. We retrieved the papers that fulfilled the research criteria, with particular attention to the second half of pregnancy, the period when most fetal surgery is performed. RESULTS: Fetuses are awake about 10% of the total time in the last gestational weeks, and they can be aroused by external stimuli. ENIn have not an anesthetic effect at normal fetal values, but only when they areartificialy injected at high doses; their blood levels in the last trimester of average pregnancies are not dissimilar either in the fetus or in the mother. CONCLUSIONS: During the second half of the pregnancy, external stimuli can awake the fetuses, although they spend most of the time in sleeping state; the presence of ENIn is absolutely not enough to guarantee an effective anesthesia during surgery. Thus, direct fetal analgesia/anesthesia is mandatory, though further studies on its possible drawbacks are necessary.


Analgesia , Fetus/surgery , Pain/embryology , Pain/prevention & control , Adenosine/blood , Adenosine/physiology , Analgesia/methods , Anesthesia/methods , Arousal , Female , Fetal Blood/chemistry , Fetus/innervation , Gestational Age , Humans , MEDLINE , Nervous System/embryology , Pain Management , Pregnancy , Pregnanolone/blood , Pregnanolone/physiology , Prostaglandin D2/blood , Prostaglandin D2/physiology
9.
Regen Med ; 13(2): 141-157, 2018 03.
Article En | MEDLINE | ID: mdl-29160149

AIM: This in vitro and in vivo study reports on silk fibroin (SF) scaffold, functionalized for in situ delivery of GABA and/or allopregnanolone (ALLO), as biomaterial for potential application in tissue engineering and nerve regeneration. MATERIALS & METHODS: We evaluated the feasibility to design 2D scaffolds (films) made of regenerated Bombyx mori SF, functionalized with GABA and/or ALLO to enhance in vitro biological functions, health, survival and growth of Schwann cells and sensitive neurons of the dorsal root ganglia. RESULTS & CONCLUSION: Our 2D-SF film showed an efficient loading and controllable release of drugs promoting nerve regeneration. SF functionalized film may be helpful for the development of bioengineered conduits and, in principle, have great potential for long-gap nerve injury repair.


Fibroins/physiology , Neurons/cytology , Schwann Cells/cytology , Silk , Tissue Engineering/methods , Animals , Biocompatible Materials , Bombyx , Fibroins/chemistry , Materials Testing , Nerve Regeneration , Pregnanolone/chemistry , Pregnanolone/physiology , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/physiology
10.
Neurosci Lett ; 649: 156-163, 2017 05 10.
Article En | MEDLINE | ID: mdl-28215878

This review highlights early efforts to translate pre-clinical and clinical findings regarding the role of neuroactive steroids in stress adaptation and PTSD into new therapeutics for PTSD. Numerous studies have demonstrated PTSD-related alterations in resting levels or the reactivity of neuroactive steroids and their targets. These studies also have demonstrated substantial variability in the dysfunction of specific neuroactive steroid systems among PTSD subpopulations. These variabilities have been related to the developmental timing of trauma, severity and type of trauma, genetic background, sex, reproductive state, lifestyle influences such as substance use and exercise, and the presence of comorbid conditions such as depression and chronic pain. Nevertheless, large naturalistic studies and a small placebo-controlled interventional study have revealed generally positive effects of glucocorticoid administration in preventing PTSD after trauma, possibly mediated by glucocorticoid receptor-mediated effects on other targets that impact PTSD risk, including other neuroactive steroid systems. In addition, clinical and preclinical studies show that administration of glucocorticoids, 17ß-estradiol, and GABAergic neuroactive steroids or agents that enhance their synthesis can facilitate extinction and extinction retention, depending on dose and timing of dose in relation to these complex PTSD-relevant recovery processes. This suggests that clinical trials designed to test neuroactive steroid therapeutics in PTSD may benefit from such considerations; typical continuous dosing regimens may not be optimal. In addition, validated and clinically accessible methods for identifying specific neuroactive steroid system abnormalities at the individual level are needed to optimize both clinical trial design and precision medicine based treatment targeting.


Steroids/physiology , Steroids/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/drug therapy , Animals , Dehydroepiandrosterone/physiology , Dehydroepiandrosterone/therapeutic use , Estradiol/physiology , Estradiol/therapeutic use , Glucocorticoids/physiology , Glucocorticoids/therapeutic use , Humans , Pregnanolone/physiology , Pregnanolone/therapeutic use , Stress Disorders, Post-Traumatic/complications , Stress, Psychological/complications
11.
Med Hypotheses ; 85(6): 947-52, 2015 Dec.
Article En | MEDLINE | ID: mdl-26365247

Impaired biosynthesis of Allopregnanolone (ALLO), a brain endogenous neurosteroid, has been associated with numerous behavioral dysfunctions, which range from anxiety- and depressive-like behaviors to aggressive behavior and changes in responses to contextual fear conditioning in rodent models of emotional dysfunction. Recent animal research also demonstrates a critical role of ALLO in social isolation. Although there are likely aspects of perceived social isolation that are uniquely human, there is also continuity across species. Both human and animal research show that perceived social isolation (which can be defined behaviorally in animals and humans) has detrimental effects on physical health, such as increased hypothalamic pituitary adrenal (HPA) activity, decreased brain-derived neurotrophic factor (BDNF) expression, and increased depressive behavior. The similarities between animal and human research suggest that perceived social isolation (loneliness) may also be associated with a reduction in the synthesis of ALLO, potentially by reducing BDNF regulation and increasing HPA activity through the hippocampus, amygdala, and bed nucleus of the stria terminalis (BNST), especially during social threat processing. Accordingly, exogenous administration of ALLO (or ALLO precursor, such as pregnenolone), in humans may help alleviate loneliness. Congruent with our hypothesis, exogenous administration of ALLO (or ALLO precursors) in humans has been shown to improve various stress-related disorders that show similarities between animals and humans i.e., post-traumatic stress disorders, traumatic brain injuries. Because a growing body of evidence demonstrates the benefits of ALLO in socially isolated animals, we believe our ALLO hypothesis can be applied to loneliness in humans, as well.


Loneliness , Pregnanolone/physiology , Amygdala/metabolism , Animals , Anxiety/psychology , Behavior, Animal/physiology , Biological Evolution , Brain-Derived Neurotrophic Factor/metabolism , Cognition , Fear/physiology , Hippocampus/metabolism , Humans , Hypothalamo-Hypophyseal System/physiology , Interpersonal Relations , Limbic System/physiology , Models, Theoretical , Pituitary-Adrenal System/physiology , Social Isolation , Stress, Psychological/physiopathology
12.
Exp Clin Psychopharmacol ; 23(1): 22-8, 2015 Feb.
Article En | MEDLINE | ID: mdl-25643026

Nicotine response may predict susceptibility to smoking relapse. Allopregnanolone, a neuroactive steroid metabolized from progesterone, has been shown to be associated with several symptoms of nicotine response. We sought to explore the association between allopregnanolone and response to nicotine during acute smoking abstinence in premenopausal women. Participants completed 2 nicotine-response laboratory sessions, 1 in their follicular (low allopregnanolone) and 1 in their luteal (high allopregnanolone) menstrual phase, on the fourth day of biochemically confirmed smoking abstinence. During the laboratory sessions, participants self-administered a nicotine nasal spray and completed a timed series of cardiovascular, cognitive, and subjective assessments of response to nicotine. The relationships of allopregnanolone with baseline values and change scores of outcome measures were assessed using covariance pattern modeling. Study participants (N = 77) had a mean age of 29.9 (SD = 6.8) years and smoked an average of 12.2 (SD = 4.9) cigarettes per day. Allopregnanolone concentration measured before nicotine administration was positively associated with systolic (ß = 0.85, p = .04) and diastolic blood pressure (ß = 1.19, p < .001) and self-report of physical symptoms (ß = 0.58, p < .001), dizziness (ß = 0.88, p < .01), jitteriness (ß = 0.90, p = .04), and pleasantness (ß = 2.05, p = .04). Allopregnanolone also had significant positive associations with change in cognition following nicotine nasal spray administration, specifically discriminability as a measure of attention (ß = 1.15, p = .05) and response bias as a measure of impulsivity (ß = 0.13, p = .02). These data suggest that allopregnanolone may be related to cardiovascular and subjective physical state during acute smoking abstinence, as well as cognitive response to nicotine.


Cognition , Pregnanolone/physiology , Premenopause , Smoking Cessation , Adolescent , Adult , Female , Humans , Smoking Cessation/psychology , Young Adult
13.
Neurosci Res ; 95: 38-50, 2015 Jun.
Article En | MEDLINE | ID: mdl-25598212

Anteroventral third ventricular region (AV3V) that regulates autonomic functions through a GABAergic mechanism possesses neuroactive steroid (NS)-synthesizing ability. Although NS can exert effects by acting on a certain type of GABAA-receptor (R), it is not clear whether NS may operate to modulate AV3V GABAergic activity for controlling autonomic functions. This study aimed to investigate the issue. AV3V infusion with a GABAA antagonist bicuculline increased plasma vasopressin (AVP), glucose, blood pressure (BP), and heart rate in rats. These events were abolished by preinjecting its agonist muscimol, whereas the infusion with allopregnanolone, a NS capable of potentiating GABAA-R function, affected none of the variables in the absence or presence of such bicuculline actions. Similarly, AV3V infusion with pregnanolone sulfate, a NS capable of antagonizing GABAA-R, produced no effect on those variables. AV3V infusion with muscimol was effective in inhibiting the responses of plasma AVP or glucose, or BP to an osmotic loading or bleeding. However, AV3V infusion with aminoglutethimide, a NS synthesis inhibitor, did not affect any of the variables in the absence or presence of those stimuli. These results suggest that NS may not cause acute effects on the AV3V GABAergic mechanism involved in regulating AVP release and other autonomic function.


Autonomic Nervous System/physiology , Pregnanolone/physiology , Prosencephalon/physiology , Receptors, GABA/physiology , Vasopressins/metabolism , Aminoglutethimide/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Autonomic Nervous System/drug effects , Bicuculline/pharmacology , Blood Glucose/drug effects , Blood Pressure/drug effects , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Heart Rate/drug effects , Male , Muscimol/pharmacology , Pregnanolone/pharmacology , Prosencephalon/drug effects , Rats , Rats, Wistar , Vasopressins/blood
14.
Fitoterapia ; 100: 35-43, 2015 Jan.
Article En | MEDLINE | ID: mdl-25447163

Gelsemium, a small genus of flowering plant from the family Loganiaceae, comprises five species including the popular Gelsemium sempervirens Ait. and Gelsemium elegans Benth., which are indigenous to North America and China/East Asia, respectively. Approximately 120 alkaloids have been isolated and identified from Gelsemium, with the predominant indole alkaloids including gelsemine, koumine, gelsemicine, gelsenicine, gelsedine, sempervirine, koumidine, koumicine and humantenine. Gelsemine is the principal active alkaloid in G. sempervirens Ait., and koumine and gelsemine are the most and second-most dominant alkaloids in G. elegans Benth. Gelsemium extract and its active alkaloids serve a variety of biological functions, including neurobiological, immunosuppressive and antitumor effects, and have traditionally been used to treat pain, neuralgia, anxiety, insomnia, asthma, respiratory ailments and cancers. This review focuses on animal-based studies of Gelsemium as a pain treatment and its mechanism of action. In contrast to morphine, when administered intrathecally and systemically, koumine, gelsemine and gelsenicine have marked antinociception in inflammatory, neuropathic and bone cancer pains without inducing antinociceptive tolerance. Gelsemium and its active alkaloids may produce antinociception by activating the spinal α3 glycine/allopregnanolone pathway. The results of this review support the clinical use of Gelsemium and suggest that its active alkaloids may be developed to treat intractable and other types of pain, preferably after chemical modification. However, Gelsemium is a known toxic plant, and its toxicity limits its appropriate dosage and clinical use. To avoid or decrease the side/toxic effects of Gelsemium, an individual monomer of highly potent alkaloids must be selected, or alkaloids that exhibit greater α3 glycine receptor selectivity may be discovered or modified.


Alkaloids/pharmacology , Analgesics/pharmacology , Gelsemium/chemistry , Pain/drug therapy , Pregnanolone/physiology , Receptors, Glycine/physiology , Animals , Indole Alkaloids/pharmacology , Mice , Molecular Structure , Rats
15.
Psychopharmacology (Berl) ; 231(17): 3537-56, 2014 Sep.
Article En | MEDLINE | ID: mdl-24997854

RATIONALE: The neurosteroid pregnenolone sulfate (PregS) acts as a cognitive enhancer and modulator of neurotransmission, yet aligning its pharmacological and physiological effects with reliable measurements of endogenous local concentrations and pharmacological and therapeutic targets has remained elusive for over 20 years. OBJECTIVES: New basic and clinical research concerning neurosteroid modulation of the central nervous system (CNS) function has emerged over the past 5 years, including important data involving pregnenolone and various neurosteroid precursors of PregS that point to a need for a critical status update. RESULTS: Highly specific actions of PregS affecting excitatory N-methyl-D-aspartate receptor (NMDAR)-mediated synaptic transmission and the pharmacological effects of PregS on various receptors and ion channels are discussed. The discovery of a high potency (nanomolar) signal transduction pathway for PregS-induced NMDAR trafficking to the cell surface via a Ca(2+)- and G protein-coupled receptor (GPCR)-dependent mechanism and a potent (EC50 ~ 2 pM) direct enhancement of intracellular Ca(2+) levels is discussed in terms of its agonist effects on long-term potentiation (LTP) and memory. Lastly, preclinical and clinical studies assessing the promnestic effects of PregS and pregnenolone toward cognitive dysfunction in schizophrenia, and altered serum levels in epilepsy and alcohol dependence, are reviewed. CONCLUSIONS: PregS is present in human and rodent brain at physiologically relevant concentrations and meets most of the criteria for an endogenous neurotransmitter/neuromodulator. PregS likely plays a significant role in modulation of glutamatergic excitatory synaptic transmission underlying learning and memory, yet the molecular target(s) for its action awaits identification.


Neuronal Plasticity/drug effects , Pregnanolone/pharmacology , Synapses/drug effects , Animals , Humans , Neurotransmitter Agents , Nootropic Agents/pharmacology , Pregnanolone/physiology , Pregnanolone/therapeutic use
16.
Prog Neurobiol ; 113: 1-5, 2014 Feb.
Article En | MEDLINE | ID: mdl-24121112

Allopregnanolone, a neuroactive steroid derived from progesterone, is synthesized within the nervous tissue, by means of specific enzymes. Contrary to progesterone and its first metabolite dihydroprogesterone, allopregnanolone is able to interact with GABA-A receptor and not with the classical progesterone receptor. This suggests that the effect of progesterone administration may be due to activation of progesterone receptor, or of GABA-A receptor, or both. However, this is rarely considered in the experimental studies. Here we summarize and discuss the hot topics involving the actions of allopregnanolone within the nervous tissue. One major role of this neuroactive steroid is neuroprotection in case of lesion, ischemia or peripheral neuropathies (i.e., diabetes). In addition, allopregnanolone may reduce the symptoms of neurodegenerative diseases (e.g., Alzheimer, Parkinson, Niemann-Pick type C, multiple sclerosis) in animal models and now translational studies are developed for its therapeutic use. Allopregnanolone may exert a beneficial effect also in case of neuropathic pain and it is also a potential candidate for the treatment of mood and anxiety disorders. Finally, this neuroactive steroid seems to have important physiological roles in the early differentiation of some neural circuits (in particular at hippocampal level), and to reduce stress during pregnancy. In conclusion, it appears that allopregnanolone is a key regulator of physiological functions and may have interesting therapeutic perspectives for neurodegenerative and psychiatric disorders.


Pregnanolone/pharmacology , Pregnanolone/physiology , Animals , Humans
17.
J Steroid Biochem Mol Biol ; 139: 144-53, 2014 Jan.
Article En | MEDLINE | ID: mdl-23669456

Neuroactive steroid concentrations are remarkably high in the foetal brain during late gestation. These concentrations are maintained by placental progesterone synthesis and the interaction of enzymes in the placenta and foetal brain. 5α-Pregnane-3α-ol-20-one (allopregnanolone) is a key neuroactive steroid during foetal life, although other 3α-hydroxy-pregnanes may make an additional contribution to neuroactive steroid action. Allopregnanolone modulates GABAergic inhibition to maintain a suppressive action on the foetal brain during late gestation. This action suppresses foetal behaviour and maintains the appropriate balance of foetal sleep-like behaviours, which in turn are important to normal neurodevelopment. Neuroactive steroid-induced suppression of excitability has a key role in protecting the foetal brain from acute hypoxia/ischaemia insults. Hypoxia-induced brain injury is markedly increased if neuroactive steroid levels are suppressed and there is increased seizure activity. There is also a rapid increase in allopregnanolone synthesis and hence levels in response to acute stress that acts as an endogenous protective mechanism. Allopregnanolone has a trophic role in regulating development, maintaining normal levels of apoptosis and increasing myelination during late gestation in the brain. In contrast, chronic foetal stressors, including intrauterine growth restriction, do not increase neuroactive steroid levels in the brain and exposure to repeated synthetic corticosteroids reduce neuroactive steroid levels. The reduced availability of neuroactive steroids may contribute to the adverse effects of chronic stressors on the foetal and newborn brain. Preterm birth also deprives the foetus of neuroactive steroid mediated protection and may increase vulnerability to brain injury and suboptimal development. These finding suggest replacement therapies should be explored. This article is part of a Special Issue entitled 'Pregnancy and steroids'.


Brain/metabolism , Fetus/metabolism , Pregnanolone/physiology , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/physiology , Animals , Brain/cytology , Brain/embryology , Female , GABAergic Neurons/metabolism , Humans , Infant, Newborn , Nerve Growth Factors/metabolism , Neuroprotective Agents/pharmacology , Placenta/enzymology , Pregnancy , Pregnanolone/pharmacology , Premature Birth/metabolism , Progesterone/physiology , Stress, Physiological
18.
Climacteric ; 16 Suppl 1: 8-17, 2013 Aug.
Article En | MEDLINE | ID: mdl-23848487

The steroidogenic endocrine glands and local synthesis both contribute to the pool of steroids present in the central nervous system and peripheral nervous system. Although the synthesis of neurosteroids in the nervous system is now well established, the spectrum of respective functions in regulating neuronal and glial functions remains to be fully elucidated. From the concept of neurosteroids derives another treatment strategy: the use of pharmaceutical agents that increase the synthesis of endogenous neurosteroids within the nervous system. This approach has so far been hampered by lack of knowledge concerning the regulation of the biosynthetic pathways of neurosteroids and their relationship with sex steroids produced by the peripheral gland or with exogenous steroids. The present review summarizes some of the available clinical and experimental findings supporting the critical role of neurosteroids during fertile life and reproductive aging and their relationship with endogenous and exogenous sex steroids. The brain metabolism of synthetic progestins and the implications of DHEA treatment in postmenopausal women will also be discussed.


Neurotransmitter Agents/physiology , Affect , Aging , Behavior , Brain Injuries , Cognition , Dehydroepiandrosterone/physiology , Dehydroepiandrosterone/therapeutic use , Dehydroepiandrosterone Sulfate/metabolism , Female , Humans , Male , Menopause , Postpartum Period/physiology , Pregnancy , Pregnanolone/physiology , Premenstrual Syndrome , Progesterone/metabolism , Progesterone/therapeutic use , Reproduction/physiology
19.
Front Neuroendocrinol ; 34(2): 132-42, 2013 Apr.
Article En | MEDLINE | ID: mdl-23563222

Parkinson's disease (PD) is associated with a massive loss of dopaminergic cells in the substantia nigra leading to dopamine hypofunction and alteration of the basal ganglia circuitry. These neurons, are under the control, among others, of the excitatory glutamatergic and inhibitory γ-aminobutyric acid (GABA) systems. An imbalance between these systems may contribute to excitotoxicity and dopaminergic cell death. Neurosteroids, a group of steroid hormones synthesized in the brain, modulate the function of several neurotransmitter systems. The substantia nigra of the human brain expresses high concentrations of allopregnanolone (3α, 5αtetrahydroprogesterone), a neurosteroid that positively modulates the action of GABA at GABAA receptors and of 5α-dihydroprogesterone, a neurosteroid acting at the genomic level. This article reviews the roles of NS acting as neuroprotectants and as GABAA receptor agonists in the physiology and pathophysiology of the basal ganglia, their impact on dopaminergic cell activity and survival, and potential therapeutic application in PD.


Dopamine/physiology , Neurotransmitter Agents/physiology , Parkinson Disease/physiopathology , Receptors, GABA-A/metabolism , Animals , Basal Ganglia/physiology , Brain/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Female , GABA-A Receptor Agonists/pharmacology , Humans , Male , Neuroprotective Agents/pharmacology , Pregnanolone/physiology , Receptors, GABA-A/drug effects , Substantia Nigra/physiology
20.
Psychoneuroendocrinology ; 38(8): 1397-406, 2013 Aug.
Article En | MEDLINE | ID: mdl-23294582

Diverse studies indicate that the alteration of the physiological levels of neurosteroids in early neonatal phases provokes alterations in the maturation of certain cerebral structures. Allopregnanolone (ALLO) has important modulatory effects in the hippocampus during the postnatal period where the adult pattern of inhibitory transmission is being established. In order to study whether endogenous neonatal ALLO levels would be a determinant parameter involved in mediating adult hippocampal GABAA system maturation, we investigated the effects of neonatal finasteride (50mg/kg, SC) treatment and ALLO (ALLO; 20mg/kg, SC) supplementation on an animal behavioural model with relevance to neurodevelopmental disorder, such as schizophrenia. Two sets of experiments were conducted. Neonatal treatment (from postnatal day (pnd) 5 to pnd9) was performed in 23 male Wistar rats and steroid quantification was performed in hippocampal homogenates at pnd9. A second group (n=127) underwent neonatal treatment (pnd5-pnd9) and were submitted to hippocampal surgery at 80d. The behavioural response to bilateral intrahippocampal neurosteroid administration (ALLO, 0.2µg/0.5µl per side or pregnenolone sulphate 5ng/0.5µl per side) on novelty-induced exploration activity and prepulse inhibition (PPI) was assessed at 95d. Results showed that neonatal ALLO and finasteride administration decreased novelty directed exploratory behaviour and impaired the prepulse inhibition of the acoustic startle response at 95 days of age. Moreover, intrahippocampal ALLO increased head-dipping behaviour independently of the neonatal treatment, while intrahippocampal ALLO decreased PPI only in finasteride and ALLO groups. The results obtained in the present study indicate the importance of neonatal neurosteroid levels in the development of hippocampal function and their relevance in a behavioural phenotype that some have likened to that present in schizophrenia.


Hippocampus/physiology , Neurotransmitter Agents/pharmacology , Pregnanolone/physiology , Sensory Gating/physiology , 5-alpha Reductase Inhibitors/pharmacology , Age Factors , Animals , Animals, Newborn , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Finasteride/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Microinjections , Motor Activity/drug effects , Neurotransmitter Agents/metabolism , Pregnanolone/administration & dosage , Pregnanolone/metabolism , Pregnanolone/pharmacology , Pregnenolone/administration & dosage , Pregnenolone/metabolism , Pregnenolone/pharmacology , Rats , Reflex, Startle/drug effects , Sensory Gating/drug effects
...