Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 445
Filter
1.
Neurosci Biobehav Rev ; 161: 105668, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608826

ABSTRACT

Neuroinflammation accompanies several brain disorders, either as a secondary consequence or as a primary cause and may contribute importantly to disease pathogenesis. Neurosteroids which act as Positive Steroid Allosteric GABA-A receptor Modulators (Steroid-PAM) appear to modulate neuroinflammation and their levels in the brain may vary because of increased or decreased local production or import from the systemic circulation. The increased synthesis of steroid-PAMs is possibly due to increased expression of the mitochondrial cholesterol transporting protein (TSPO) in neuroinflammatory tissue, and reduced production may be due to changes in the enzymatic activity. Microglia and astrocytes play an important role in neuroinflammation, and their production of inflammatory mediators can be both activated and inhibited by steroid-PAMs and GABA. What is surprising is the finding that both allopregnanolone, a steroid-PAM, and golexanolone, a novel GABA-A receptor modulating steroid antagonist (GAMSA), can inhibit microglia and astrocyte activation and normalize their function. This review focuses on the role of steroid-PAMs in neuroinflammation and their importance in new therapeutic approaches to CNS and liver disease.


Subject(s)
Neuroinflammatory Diseases , Pregnanolone , Pregnanolone/pharmacology , Pregnanolone/metabolism , Humans , Animals , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Microglia/drug effects , Microglia/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , GABA-A Receptor Antagonists/pharmacology
2.
J Steroid Biochem Mol Biol ; 241: 106525, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636682

ABSTRACT

Gamma-aminobutyric acid A (GABA-A) receptors in the cells of the immune system enhance anti-inflammatory responses by regulating cytokine secretion, cytotoxic responses, and cell activation. In the CNS, the formation of GABA-A subunits into a pentameric structure has been extensively studied; however, no such study has been conducted in the immune system. The objective of the present study was to examine associations between the levels of steroid hormones and GABA-A receptor δ subunit expression in the immune system. We focused on this subunit because GABA-A receptors that contain it become significantly more sensitive to steroid hormones. We collected 80 blood samples from reproductive age women for the purpose of analyzing dehydroepiandrosterone (DHEA), 17ß-estradiol, progesterone, and allopregnanolone using liquid chromatography-mass spectrometry (LC-MS). Furthermore, we extracted peripheral blood mononuclear cells (PBMCs) for determining mRNA expression levels of GABA-A receptor genes encoding the δ and ε subunits. We constructed linear mixed effect models for each GABA-A receptor subunit with all 4 steroid hormones, age, and age of menarche as predictors. Whereas DHEA was significantly associated with δ subunit expression (t-value = 2.981; p = 0.003), in line with our hypothesis, none of the steroid hormones were significantly associated with the expression of the ε subunit. Results of this study indicate that significant interactions between hormones from the steroid hormone biosynthesis pathway and GABAergic machinery from the immune cells may be utilized to expand models examining the molecular basis of inflammatory conditions.


Subject(s)
Dehydroepiandrosterone , Receptors, GABA-A , Humans , Female , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Adult , Progesterone/blood , Young Adult , Estradiol/blood , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Pregnanolone/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Expression/drug effects
3.
Biomolecules ; 14(4)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38672476

ABSTRACT

The recent approval of formulations of the endogenous neurosteroid allopregnanolone (brexanolone) and the synthetic neuroactive steroid SAGE-217 (zuranolone) to treat postpartum depression (PPD) has encouraged further research to elucidate why these potent enhancers of GABAAR function are clinically effective in this condition. Dopaminergic projections from the ventral tegmental area (VTA) to the nucleus accumbens are associated with reward/motivation and brain imaging studies report that individuals with PPD show reduced activity of this pathway in response to reward and infant engagement. However, the influence of neurosteroids on GABA-ergic transmission in the nucleus accumbens has received limited attention. Here, we investigate, in the medium spiny neurons (MSNs) of the mouse nucleus accumbens core, the effect of allopregnanolone, SAGE-217 and other endogenous and synthetic steroids of interest on fast phasic and tonic inhibition mediated by synaptic (α1/2ßγ2) and extrasynaptic (α4ßδ) GABAARs, respectively. We present evidence suggesting the resident tonic current results from the spontaneous opening of δ-GABAARs, where the steroid-enhanced tonic current is GABA-dependent. Furthermore, we demonstrate local neurosteroid synthesis in the accumbal slice preparation and reveal that GABA-ergic neurotransmission of MSNs is influenced by an endogenous neurosteroid tone. Given the dramatic fluctuations in allopregnanolone levels during pregnancy and postpartum, this neurosteroid-mediated local fine-tuning of GABAergic transmission in the MSNs will probably be perturbed.


Subject(s)
Neurosteroids , Nucleus Accumbens , Pregnanolone , Receptors, GABA-A , Animals , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Mice , Receptors, GABA-A/metabolism , Neurosteroids/metabolism , Pregnanolone/pharmacology , Pregnanolone/metabolism , Synapses/metabolism , Synapses/drug effects , Mice, Inbred C57BL , Female , Male , Synaptic Transmission/drug effects , Neurons/metabolism , Neurons/drug effects
4.
Adv Med Sci ; 69(1): 176-189, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38561071

ABSTRACT

PURPOSE: Metabolic syndrome (MetS) is a common disorder associated with disturbed neurotransmitter homeostasis. Memantine, an N-methyl-d-aspartate receptor (NMDAR) antagonist, was first used in Alzheimer's disease. Allopregnanolone (Allo), a potent positive allosteric modulator of the Gamma-Amino-Butyric Acid (GABA)-A receptors, decreases in neurodegenerative diseases. The study investigated the impact of Memantine versus Allo administration on the animal model of MetS to clarify whether the mechanism of abnormalities is related more to excitatory or inhibitory neurotransmitter dysfunction. MATERIALS AND METHODS: Fifty-six male rats were allocated into 7 groups: 4 control groups, 1 MetS group, and 2 treated MetS groups. They underwent assessment of cognition-related behavior by open field and forced swimming tests, electroencephalogram (EEG) recording, serum markers confirming the establishment of MetS model and hippocampal Glial Fibrillary Acidic Protein (GFAP) and Brain-Derived Neurotrophic Factor (BDNF). RESULTS: Allo improved anxiety-like behavior and decreased grooming frequency compared to Memantine. Both drugs increased GFAP and BDNF expression, improving synaptic plasticity and cognition-related behaviors. The therapeutic effect of Allo was more beneficial regarding lipid profile and anxiety. We reported progressive slowing of EEG waves in the MetS group with Memantine and Allo treatment with increased relative theta and decreased relative delta rhythms. CONCLUSIONS: Both Allo and Memantine boosted the outcome parameters in the animal model of MetS. Allo markedly improved the anxiety-like behavior in the form of significantly decreased grooming frequency compared to the Memantine-treated groups. Both drugs were associated with increased hippocampal GFAP and BDNF expression, indicating an improvement in synaptic plasticity and so, cognition-related behaviors.


Subject(s)
Memantine , Metabolic Syndrome , Neuronal Plasticity , Receptors, GABA-A , Receptors, N-Methyl-D-Aspartate , Animals , Neuronal Plasticity/drug effects , Male , Rats , Metabolic Syndrome/metabolism , Metabolic Syndrome/drug therapy , Receptors, N-Methyl-D-Aspartate/metabolism , Memantine/pharmacology , Receptors, GABA-A/metabolism , Brain/metabolism , Brain/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Pregnanolone/pharmacology , Pregnanolone/metabolism , Rats, Wistar , Disease Models, Animal
5.
J Neuroendocrinol ; 36(4): e13378, 2024 04.
Article in English | MEDLINE | ID: mdl-38482748

ABSTRACT

Neurosteroids have been implicated in the pathophysiology of post-traumatic stress disorder (PTSD). Allopregnanolone is reduced in subsets of individuals with PTSD and has been explored as a novel treatment strategy. Both direct trauma exposure and witnessed trauma are risk factors for PTSD; however, the role of neurosteroids in the behavioral outcomes of these unique experiences has not been explored. Here, we investigate whether observational fear is associated with a reduced capacity for endogenous neurosteroidogenesis and the relationship with behavioral outcomes. We demonstrated that mice directly subjected to a threat (foot shocks) and those witnessing the threat have decreased plasma levels of allopregnanolone. The expression of a key enzyme involved in endogenous neurosteroid synthesis, 5α-reductase type 2, is decreased in the basolateral amygdala, which is a major emotional processing hub implicated in PTSD. We demonstrated that genetic knockdown or pharmacological inhibition of 5α-reductase type 2 exaggerates the behavioral expression of fear in response to witnessed trauma, whereas oral treatment with an exogenous, synthetic neuroactive steroid gamma-aminobutyric acid-A receptor positive allosteric modulator with molecular pharmacology similar to allopregnanolone (SGE-516 [tool compound]) decreased the behavioral response to observational fear. These data implicate impaired endogenous neurosteroidogenesis in the pathophysiology of threat exposure, both direct and witnessed. Further, these data suggest that treatment with exogenous 5α-reduced neurosteroids or targeting endogenous neurosteroidogenesis may be beneficial for the treatment of individuals with PTSD, whether resulting from direct or witnessed trauma.


Subject(s)
Neurosteroids , Animals , Mice , Pregnanolone/metabolism , Receptors, GABA-A/metabolism , Fear/physiology , Emotions , Cholestenone 5 alpha-Reductase/metabolism
6.
Neurosci Biobehav Rev ; 158: 105558, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38244954

ABSTRACT

This mini-review presents emerging evidence that endogenous neurosteroids modulate both pro- and anti-inflammatory signaling by immune cells and brain cells that contribute to depression, alcohol use disorders, and other inflammatory conditions. We first review the literature on pregnenolone and allopregnanolone inhibition of proinflammatory neuroimmune pathways in the periphery and the brain - effects that are independent of GABAergic mechanisms. We follow with evidence for neurosteroid enhancement of anti-inflammatory and protective pathways in brain and immune cells. These studies draw clinical relevance from a large body of evidence that pro-inflammatory immune signaling is dysregulated in many brain disorders and the fact that neurosteroids inhibit the same inflammatory pathways that are activated in depression, alcohol use disorders and other inflammatory conditions. Thus, we describe evidence that neurosteroid levels are decreased and neurosteroid supplementation has therapeutic efficacy in these neuropsychiatric conditions. We conclude with a perspective that endogenous regulation of immune balance between pro- and anti-inflammatory pathways by neurosteroid signaling is essential to prevent the onset of disease. Deficits in neurosteroids may unleash excessive pro-inflammatory activation which progresses in a feed-forward manner to disrupt brain networks that regulate stress, emotion and motivation. Neurosteroids can block various inflammatory pathways in mouse and human macrophages, rat brain and human blood and therefore provide new hope for treatment of intractable conditions that involve excessive inflammatory signaling.


Subject(s)
Alcoholism , Neurosteroids , Rats , Humans , Mice , Animals , Neurosteroids/metabolism , Alcoholism/metabolism , Brain/metabolism , Pregnanolone/pharmacology , Pregnanolone/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
7.
Asian J Psychiatr ; 90: 103800, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37898099

ABSTRACT

To reveal the exact changes of allopregnanolone-mediated γ-aminobutyric acid A receptor pathways and its specific therapeutic targets by Shuyu Capsule treating premenstrual depression, female Wistar rat models of premenstrual depression was established by Forced swimming test (FST). Behavioral tests, enzyme-linked immunosorbent assay (ELISA), interference knockdown adenovirus, and overexpressed vector adenovirus of GABAARδ, RT-qPCR, Western-Blot, and immunohistochemical detecting expressions were applied to identify the therapeutic targets. FST-based rat model indicated that Shuyu capsules alleviated typical premenstrual depression and may regulate alternations of 5α-reductase and 3α-steroid dehydrogenase, enhancing the metabolic pathway of progesterone to allopregnanolone, as well as targeting the GABAARδ subunit, thereby alleviating premenstrual depression of PMDD rat models.


Subject(s)
Pregnanolone , Receptors, GABA-A , Rats , Female , Animals , Rats, Wistar , Pregnanolone/metabolism , Pregnanolone/therapeutic use , Hippocampus/metabolism , gamma-Aminobutyric Acid , Metabolic Networks and Pathways
8.
Psychoneuroendocrinology ; 156: 106327, 2023 10.
Article in English | MEDLINE | ID: mdl-37423029

ABSTRACT

BACKGROUND: Anxiety disorders are the most common psychiatric disorder during the perinatal period and one of the major risk factors for postpartum depression, yet we know little about biological factors in the etiology of perinatal anxiety. A growing literature points to neuroactive steroid (NAS) dysregulation in perinatal mental illness, but directionality has not been clearly demonstrated, results are not consistent, and no studies have investigated NAS in a population with pure anxiety without comorbid depression. We aimed to add to the limited literature by examining the association between anxiety without comorbid depression and metabolic pathways of NAS longitudinally across the peripartum. METHODS: We measured anxiety symptoms by psychological scales and NAS levels using Gas Chromatography-Mass Spectrometry (GC-MS) at the second and third trimester (T2 and T3) and week 6 postpartum (W6) in n = 36 women with anxiety and n = 38 healthy controls. The anxiety group was determined by a data-driven approach, and cross-sectional and longitudinal statistical methods were used to examine the relationship between the study population and NAS. RESULTS: We found that anxiety had a significant moderating effect on the relationship between progesterone and allopregnanolone, with no such effect for the relationships between progesterone and the intermediate (5α-DHP) or isomeric (isoallopregnanolone) compounds in this pathway, and no effects on the corresponding pathway converting progesterone to pregnanolone and epipregnanolone. We also found a less precipitous decline in the ratio of allopregnanolone to progesterone between T3 and W6 in the anxiety group compared to the non-anxiety group. A genotype analysis of a single-nucleotide polymorphism in the AKR1C2 gene demonstrated that the relationship of allopregnanolone to the intermediate metabolite, 5α-DHP, differed by genotype. CONCLUSION: Our exploratory findings indicate that, for pregnant people with anxiety, metabolism is shunted more aggressively toward the endpoint of the progesterone to allopregnanolone metabolic pathway than it is for those without anxiety.


Subject(s)
Neurosteroids , Progesterone , Pregnancy , Humans , Female , Progesterone/metabolism , 5-alpha-Dihydroprogesterone , Pregnanolone/metabolism , Cross-Sectional Studies , Anxiety Disorders
9.
Genes (Basel) ; 14(6)2023 06 08.
Article in English | MEDLINE | ID: mdl-37372414

ABSTRACT

Brexanolone, a formulation of the neurosteroid allopregnanolone (ALLO), is approved for treating postpartum depression (PPD) and is being investigated for therapeutic efficacy across numerous neuropsychiatric disorders. Given ALLO's beneficial effects on mood in women with PPD compared to healthy control women, we sought to characterize and compare the cellular response to ALLO in women with (n = 9) or without (n = 10, i.e., Controls) past PPD, utilizing our previously established patient-derived lymphoblastoid cell lines (LCLs). To mimic in vivo PPD ALLO-treatment, LCLs were exposed to ALLO or DMSO vehicle for 60 h and RNA-sequenced to detect differentially expressed genes (DEGs, pnominal < 0.05). Between ALLO-treated Control and PPD LCLs, 269 DEGs were identified, including Glutamate Decarboxylase 1 (GAD1), which was decreased 2-fold in PPD. Network analysis of PPD:ALLO DEGs revealed enriched terms related to synaptic activity and cholesterol biosynthesis. Within-diagnosis analyses (i.e., DMSO vs. ALLO) detected 265 ALLO-induced DEGs in Control LCLs compared to only 98 within PPD LCLs, with just 11 DEGs overlapping. Likewise, the gene ontologies underlying ALLO-induced DEGs in PPD and Control LCLs were divergent. These data suggest that ALLO may activate unique and opposing molecular pathways in women with PPD, which may be tied to its antidepressant mechanism.


Subject(s)
Depression, Postpartum , Pregnanolone , Humans , Female , Pregnanolone/pharmacology , Pregnanolone/metabolism , Pregnanolone/therapeutic use , Depression, Postpartum/drug therapy , Depression, Postpartum/genetics , Depression, Postpartum/metabolism , Transcriptome/genetics , Dimethyl Sulfoxide , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use
10.
J Endocrinol ; 258(1)2023 07 01.
Article in English | MEDLINE | ID: mdl-37115241

ABSTRACT

Neuroactive steroids can rapidly regulate multiple physiological functions in the central and peripheral nervous systems. The aims of the present study were to determine whether allopregnanolone (ALLO), administered in low nanomolar and high micromolar concentrations, can: (i) induce changes in the ovarian progesterone (P4) and estradiol (E2) release; (ii) modify the ovarian mRNA expression of Hsd3b1 (3ß-hydroxysteroid dehydrogenase, 3ß-HSD)3ß-, Akr1c3 (20α-hydroxysteroid dehydrogenase, 20α-HSD), and Akr1c14 (3α-hydroxy steroid oxidoreductase, 3α-HSOR)); and (iii) modulate the ovarian expression of progesterone receptors A and B, α and ß estrogenic receptors, luteinizing hormone receptor (LHR) and follicle-stimulating hormone receptor (FSHR). To further characterize ALLO peripheral actions, the effects were evaluated using a superior mesenteric ganglion-ovarian nervous plexus-ovary (SMG-ONP-O) and a denervated ovary (DO) systems. ALLO SMG administration increased P4 concentration in the incubation liquid by decreasing ovarian 20α-HSD mRNA, and it also increased ovarian 3α-HSOR mRNA expression. In addition, ALLO neural peripheral modulation induced an increase in the expression of ovarian LHR, PRA, PRB, and ERα. Direct ALLO administration to the DO decreased E2 and increased P4 concentration in the incubation liquid. The mRNA expression of 3ß-HSD decreased and 20α-HSD increased. Further, ALLO in the OD significantly changed ovarian FSHR and PRA expression. This is the first evidence of ALLO's direct effect on ovarian steroidogenesis. Our results provide important insights about how this neuroactive steroid interacts both with the PNS and the ovary, and these findings might help devise some of the pleiotropic effects of neuroactive steroids on female reproduction. Moreover, ALLO modulation of ovarian physiology might help uncover novel treatment approaches for reproductive diseases.


Subject(s)
Neurosteroids , Pregnanolone , Female , Humans , Pregnanolone/pharmacology , Pregnanolone/metabolism , Neurosteroids/metabolism , Neurosteroids/pharmacology , Ovary/metabolism , Progesterone/pharmacology , Progesterone/metabolism , Hydroxysteroid Dehydrogenases/metabolism , Hydroxysteroid Dehydrogenases/pharmacology , RNA, Messenger/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/pharmacology
11.
Aging (Albany NY) ; 15(4): 1143-1157, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36842096

ABSTRACT

BACKGROUND: Premenstrual dysphoric disorder (PMDD) is a common mental health challenge among women of reproductive age. Allopregnanolone (3α, 5α-THP; ALLO) mediated functional alterations of GABAA receptors (GABAA-R) are involved in PMDD pathogenesis, however, the specific mechanism remains unknown. Therefore, we investigated the role of ALLO mediated GABAA-Rα4 in the pathophysiology of PMDD. PURPOSE: We determined whether the pathogenesis of PMDD is associated with ALLO mediated GABAA-Rα4 expression changes in different brain regions. METHODS: Rat models of PMDD liver-qi invasion syndrome (PMDD-LIS) were established via the resident intruder paradigm. Behavioral changes of rats were assessed by aggressive behavior tests, EPM and OFT. The levels of progesterone and ALLO in serum as well as brain areas were determined by ELISA. Variations in GABAA-Rα4 levels in brain regions were assessed by immunofluorescence and RT-PCR. Medicated serum was used to interfere with rat hippocampal neurons, and changes in Cl- current were recorded through electrophysiology. RESULTS: Premenstrual anxiety and irritability of PMDD-LIS patients can be simulated in PMDD-LIS rat models. Exogenous ALLO significantly improved the anxiety behaviors of PMDD-LIS rats. Changes in ALLO among different brain regions varied. GABAA-Rα4 expressions were low in the amygdala and abnormally high in the hippocampus, however, ALLO alleviated these deviations. Whole-cell patch clamp recording technique showed a weaker Cl- current intensity of PMDD-LIS rats, reduced neuroinhibitory functions and increased Cl- current intensity in the ALLO group drug serum intervention and enhanced emotional inhibition function. CONCLUSION: We established that ALLO regulation of the GABAA-Rα4 subunit in the amygdala and hippocampus is involved in PMDD-LIS pathogenesis.


Subject(s)
Liver Diseases , Premenstrual Dysphoric Disorder , Humans , Female , Rats , Animals , Pregnanolone/pharmacology , Pregnanolone/metabolism , Qi , Hippocampus/metabolism , Amygdala/metabolism
12.
Alcohol Clin Exp Res (Hoboken) ; 47(3): 459-469, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36587947

ABSTRACT

BACKGROUND: Alcohol affects multiple circuits in the brain, mainly disrupting the delicate balance between inhibitory γ-aminobutyric acid (GABA) transmission and excitatory glutamate signaling in brain areas involved in reward circuits. These include the amygdala, nucleus accumbens (Acb), and ventral tegmental area (VTA). This action impairs circuits that regulate behavioral control of craving and alcohol seeking and intake. Studies in both rodent models and postmortem human brain of patients with alcohol use disorder (AUD) have highlighted the association between the loss of GABAergic inhibition and the development of addiction. The neurosteroid (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP) is a potent positive modulator of GABAA receptors. Chronic alcohol consumption reduces 3α,5α-THP levels, resulting in decreased GABA inhibition. We previously demonstrated that enhancing neurosteroid biosynthesis by overexpression of the cholesterol side-chain cleavage enzyme P450scc decreased alcohol intake in male alcohol-preferring rats (P-rats). While most of the evidence of alcohol-induced alterations comes from studies in male subjects, some data show that females are more vulnerable to alcohol's effects than males. METHODS: In this study, we investigated the ability of 3α,5α-THP direct infusions in two brain regions that contribute to alcohol reinforcement, the VTA and Acb core (AcbC), to regulate alcohol self-administration in female P-rats. RESULTS: Administration of 3α,5α-THP into the AcbC increased 3α,5α-THP-positive cell expression in this area and reduced alcohol self-administration. By contrast, 3α,5α-THP infusion into the VTA did not significantly affect alcohol self-administration, though trends for a reduction were found. CONCLUSIONS: Our results show that local increases in 3α,5α-THP in the AcbC may alter mesolimbic activity that drives a reduction in alcohol self-administration.


Subject(s)
Neurosteroids , Nucleus Accumbens , Humans , Rats , Male , Female , Animals , Nucleus Accumbens/metabolism , Neurosteroids/metabolism , Neurosteroids/pharmacology , Ethanol , Brain , Pregnanolone/metabolism , gamma-Aminobutyric Acid/metabolism
13.
J Neuroendocrinol ; 35(2): e13197, 2023 02.
Article in English | MEDLINE | ID: mdl-36165431

ABSTRACT

Rats can use several memory systems to navigate a maze toward a reward. Two of these are place memory and response memory and female rats can be biased to predominantly use one over another. Both progesterone and estrogens have been shown to alter memory bias. Although the effects of estrogens have been well documented, the effects of progesterone remain somewhat unexplored. Mechanisms through which progesterone may be acting to exert its effects are reviewed here. Converging evidence suggests that the actions of progesterone differ depending on the presence of estrogens, frequently acting in opposition to estrogens when administered together. The hippocampus, dorsal striatum, and prefrontal cortex are likely involved, as is the progesterone metabolite, allopregnanolone. There is a need for more research on progesterone and memory bias, especially considering current formulations of hormonal contraceptives include progestins.


Subject(s)
Progesterone , Spatial Navigation , Female , Rats , Animals , Progesterone/metabolism , Progestins/pharmacology , Estrogens/metabolism , Pregnanolone/metabolism , Spatial Memory , Maze Learning , Hippocampus/metabolism
14.
Nat Commun ; 13(1): 7537, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36476469

ABSTRACT

Early-life adversity (ELA) increases the likelihood of neuropsychiatric diagnoses, which are more prevalent in women than men. Since changes in reproductive hormone levels can also increase the probability of anxiety disorders in women, we examined the effects of ELA on adult female mice across the estrous cycle. We found that during diestrus, when progesterone levels are relatively high, ELA mice exhibit increased avoidance behavior and increased theta oscillation power in the ventral hippocampus (vHIP). We also found that diestrus ELA mice had higher levels of progesterone and lower levels of allopregnanolone, a neurosteroid metabolite of progesterone, in the vHIP compared with control-reared mice. Progesterone receptor antagonism normalized avoidance behavior in ELA mice, while treatment with a negative allosteric modulator of allopregnanolone promoted avoidance behavior in control mice. These results suggest that altered vHIP progesterone and allopregnanolone signaling during diestrus increases avoidance behavior in ELA mice.


Subject(s)
Avoidance Learning , Estrous Cycle , Progesterone , Animals , Female , Mice , Progesterone/metabolism , Pregnanolone/metabolism
15.
Pharmacol Ther ; 240: 108299, 2022 12.
Article in English | MEDLINE | ID: mdl-36323379

ABSTRACT

Alcohol Use Disorder (AUD) is a multifaceted relapsing disorder that is commonly comorbid with psychiatric disorders, including anxiety. Alcohol exposure produces a plethora of effects on neurobiology. Currently, therapeutic strategies are limited, and only a few treatments - disulfiram, acamprosate, and naltrexone - are available. Given the complexity of this disorder, there is a great need for the identification of novel targets to develop new pharmacotherapy. The GABAergic system, the primary inhibitory system in the brain, is one of the well-known targets for alcohol and is responsible for the anxiolytic effects of alcohol. Interestingly, GABAergic neurotransmission is fine-tuned by neuroactive steroids that exert a regulatory role on several endocrine systems involved in neuropsychiatric disorders including AUD. Mounting evidence indicates that alcohol alters the biosynthesis of neurosteroids, whereas acute alcohol increases and chronic alcohol decreases allopregnanolone levels. Our recent work highlighted that chronic alcohol-induced changes in neurosteroid levels are mediated by epigenetic modifications, e.g., DNA methylation, affecting key enzymes involved in neurosteroid biosynthesis. These changes were associated with changes in GABAA receptor subunit expression, suggesting an imbalance between excitatory and inhibitory signaling in AUD. This review will recapitulate the role of neurosteroids in the regulation of the neuroendocrine system, highlight their role in the observed allostatic load in AUD, and develop a framework from mechanisms to potential pharmacotherapy.


Subject(s)
Alcoholism , Neurosteroids , Humans , Pregnanolone/metabolism , Alcoholism/drug therapy , Receptors, GABA-A/metabolism , Anxiety , Ethanol
16.
Acta Neurobiol Exp (Wars) ; 82(3): 347-357, 2022.
Article in English | MEDLINE | ID: mdl-36214717

ABSTRACT

Postpartum depression (PPD) is the most common type of puerperal mental syndrome and affects maternal physical and mental health and even the growth and development of infants. Paeoniflorin exerts a potential antidepressive effect; however, the functional roles and potential mechanisms of paeoniflorin in PPD are still largely unknown. PPD rat models were prepared by withdrawing hormone­simulated pregnancy (HSP), and subjects were treated with paeoniflorin and fluoxetine or plasmids. The sucrose preference test (SPT), forced swimming test (FST) and tail suspension test (TST) were used to monitor depression­like behavior in rats. A radioimmunoassay was utilized for estradiol (E2) and progesterone (P) measurements. ELISA was performed to detect serum corticosterone (Cor), hippocampal allopregnanolone (Allo), IL­1ß and TNF­α levels. Expression of the E2 receptors ERα and ERß was detected by qPCR. Western blotting was used to detect TSPO, BDNF and mTOR phosphorylation. Paeoniflorin drastically increased the sucrose preference of rats while decreasing the immobility time in the FST and TST in PPD models. Moreover, paeoniflorin intervention upregulated serum E2, hippocampal Allo, ERα, and ERß levels but degraded P, serum Cor, IL­1ß, TNF­α and ERα/ERß levels. Mechanistically, paeoniflorin promoted TSPO and BDNF­mTOR pathway activation in PPD rats. Furthermore, suppression of TSPO or the BDNF­mTOR pathway partially reversed the effects of paeoniflorin on depression­like behaviors, hormone levels, and inflammatory cytokine release. Paeoniflorin may improve symptoms of PPD by regulating the TSPO and BDNF­mTOR pathways, indicating that paeoniflorin may be an effective anti­PPD and antidepressant drug, providing evidence for the future treatment of PPD.


Subject(s)
Brain-Derived Neurotrophic Factor , Depression, Postpartum , Glucosides , Monoterpenes , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Carrier Proteins/metabolism , Corticosterone/metabolism , Depression/drug therapy , Depression, Postpartum/drug therapy , Depression, Postpartum/metabolism , Disease Models, Animal , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Fluoxetine/therapeutic use , Glucosides/pharmacology , Hippocampus/metabolism , Humans , Monoterpenes/pharmacology , Pregnancy , Pregnanolone/metabolism , Progesterone/metabolism , Rats , Receptors, GABA-A/metabolism , Stress, Psychological , Sucrose , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Aging (Albany NY) ; 14(20): 8437-8447, 2022 10 24.
Article in English | MEDLINE | ID: mdl-36287173

ABSTRACT

BACKGROUND: Premenstrual dysphoric disorder (PMDD) is a severe mood disorder with pathological changes rooted in GABRB2 copy number variation. Here, we aimed to elucidate the gene dose effect and allopregnanolone binding mechanism of Gabrb2 on possible PMDD-like and comorbid phenotypes in knockout mice. METHODS: PMDD-like behaviors of Gabrb2-knockout mice were measured through various tests. Western Blot and ELISA were used to detect changes in the GABAAR subunits and related neurotransmitter changes in mice respectively for the internal mechanism. The response of mice to allopregnanolone (ALLO) was examined through an exogenous ALLO injection, then validated by the patch-clamp technique to elaborate the potential mechanism of ALLO-mediated GABAAR. RESULTS: Gabrb2-knockout mice displayed changes in anxiety-like and depression-like emotions opposite to PMDD symptoms, changes in social, learning, and memory capacities similar to PMDD symptoms, and pain threshold changes opposite to PMDD symptoms. GABAAR δ subunit expression in the brains of the Gabrb2-knockout mice was significantly higher than that of Wild-type mice (P<0.05). Gabrb2-knockout mice demonstrated neurotransmitter metabolism disturbance of GABA, Glu, acetylcholine, DA, norepinephrine, and epinephrine. Moreover, Gabrb2-knockout mice did not display the expected phenotypic effect after ALLO injection. Relative to WT mice, the knockout of the ß2 subunit gene enhanced the agonistic effect of ALLO on GABAA receptors in cortical neuronal cells. CONCLUSIONS: GABAAR ß 2 regulates PMDD-like behaviors. The ALLO binding site may not be located on ß two subunits, abnormal δ and ε subunit expression in the mouse brain and the disturbance of neurotransmitters may result in ALLO sensitivity.


Subject(s)
Premenstrual Dysphoric Disorder , Humans , Female , Animals , Mice , Pregnanolone/metabolism , Receptors, GABA-A/genetics , Mice, Knockout , DNA Copy Number Variations
18.
Int J Mol Sci ; 23(9)2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35563388

ABSTRACT

Glioblastomas (GBs) are the most aggressive and common primary malignant brain tumors. Steroid hormone progesterone (P4) and its neuroactive metabolites, such as allopregnanolone (3α-THP) are synthesized by neural, glial, and malignant GB cells. P4 promotes cellular proliferation, migration, and invasion of human GB cells at physiological concentrations. It has been reported that 3α-THP promotes GB cell proliferation. Here we investigated the effects of 3α-THP on GB cell migration and invasion, the participation of the enzymes involved in its metabolism (AKR1C1-4), and the role of the c-Src kinase in 3α-THP effects in GBs. 3α-THP 100 nM promoted migration and invasion of U251, U87, and LN229 human-derived GB cell lines. We observed that U251, LN229, and T98G cell lines exhibited a higher protein content of AKR1C1-4 than normal human astrocytes. AKR1C1-4 silencing did not modify 3α-THP effects on migration and invasion. 3α-THP activated c-Src protein at 10 min (U251 cells) and 15 min (U87 and LN229 cells). Interestingly, the pharmacological inhibition of c-Src decreases the promoting effects of 3α-THP on cell migration and invasion. Together, these data indicate that 3α-THP promotes GB migration and invasion through c-Src activation.


Subject(s)
CSK Tyrosine-Protein Kinase , Glioblastoma , Pregnanolone , CSK Tyrosine-Protein Kinase/metabolism , Cell Proliferation , Glioblastoma/metabolism , Humans , Pregnanolone/metabolism , Pregnanolone/pharmacology , Protein-Tyrosine Kinases
19.
J Psychopharmacol ; 36(4): 439-448, 2022 04.
Article in English | MEDLINE | ID: mdl-35475379

ABSTRACT

BACKGROUND: Anxiety disorders are highly prevalent affecting up to 33.7% of people over a lifetime. Although many treatment options are available, they are often associated with unacceptable side-effect profiles and approximately one in three patients are treatment resistant. Allopregnanolone, a neuroactive steroid acting as a positive allosteric modulator at the GABAA receptor, is synthesised in response to stress and acts to negatively modulate the hypothalamic-pituitary-adrenal axis. FINDINGS: After chronic exposure to and withdrawal from allopregnanolone, an increase in α4ß2δ GABAA receptors results in a reduced inhibitory effect of allopregnanolone, resulting in decreased inhibition and, therefore, increased neuronal excitability. The relationship between allopregnanolone and increased α4ß2δ GABAA receptors has been demonstrated in animal models during methamphetamine withdrawal and puberty, events both associated with stress. The effect of allopregnanolone during these events is anxiogenic, a paradoxical action to its usual anxiolytic effects. Flumazenil, the GABAA receptor antagonist, has been shown to cause receptor internalisation of α4ß2δ GABAA receptors, which may results in anxiolysis. CONCLUSION: We propose that chronic stress and chronic exposure to and withdrawal from allopregnanolone in anxiety disorders result in alterations in GABAA receptor function, which can be corrected by flumazenil. As such, flumazenil may exhibit anxiolytic properties in patients with increased α4ß2δ GABAA receptor expression.


Subject(s)
Anti-Anxiety Agents , Flumazenil , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety Disorders/drug therapy , Flumazenil/pharmacology , Humans , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Pregnanolone/metabolism , Pregnanolone/pharmacology , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid
20.
Cell Mol Neurobiol ; 42(1): 23-40, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34138412

ABSTRACT

Progesterone regulates a number of processes in neurons and glial cells not directly involved in reproduction or sex behavior. Several neuroprotective effects are better observed under pathological conditions, as shown in the Wobbler mouse model of amyotrophic laterals sclerosis (ALS). Wobbler mice are characterized by forelimb atrophy due to motoneuron degeneration in the spinal cord, and include microgliosis and astrogliosis. Here we summarized current evidence on progesterone reversal of Wobbler neuropathology. We demonstrated that progesterone decreased motoneuron vacuolization with preservation of mitochondrial respiratory complex I activity, decreased mitochondrial expression and activity of nitric oxide synthase, increased Mn-dependent superoxide dismutase, stimulated brain-derived neurotrophic factor, increased the cholinergic phenotype of motoneurons, and enhanced survival with a concomitant decrease of death-related pathways. Progesterone also showed differential effects on glial cells, including increased oligodendrocyte density and downregulation of astrogliosis and microgliosis. These changes associate with reduced anti-inflammatory markers. The enhanced neurochemical parameters were accompanied by longer survival and increased muscle strength in tests of motor behavior. Because progesterone is locally metabolized to allopregnanolone (ALLO) in nervous tissues, we also studied neuroprotection by this derivative. Treatment of Wobbler mice with ALLO decreased oxidative stress and glial pathology, increased motoneuron viability and clinical outcome in a progesterone-like manner, suggesting that ALLO could mediate some progesterone effects in the spinal cord. In conclusion, the beneficial effects observed in different parameters support the versatile properties of progesterone and ALLO in a mouse model of motoneuron degeneration. The studies foresee future therapeutic opportunities with neuroactive steroids for deadly diseases like ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Neuroprotective Agents , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Mice , Motor Neurons , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Pregnanolone/metabolism , Pregnanolone/pharmacology , Pregnanolone/therapeutic use , Progesterone/metabolism , Progesterone/pharmacology , Progesterone/therapeutic use , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...