Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 22(11)2021 May 23.
Article in English | MEDLINE | ID: mdl-34071053

ABSTRACT

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.


Subject(s)
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
2.
J Lipid Res ; 61(10): 1308-1319, 2020 10.
Article in English | MEDLINE | ID: mdl-32669383

ABSTRACT

Cognitive decline with age is a harmful process that can reduce quality of life. Multiple factors have been established to contribute to cognitive decline, but the overall etiology remains unknown. Here, we hypothesized that cognitive dysfunction is mediated, in part, by increased levels of inflammatory cytokines that alter allopregnanolone (AlloP) levels, an important neurosteroid in the brain. We assessed the levels and regulation of AlloP and the effects of AlloP supplementation on cognitive function in 4-month-old and 24-month-old male C57BL/6 mice. With age, the expression of enzymes involved in the AlloP synthetic pathway was decreased and corticosterone (CORT) synthesis increased. Supplementation of AlloP improved cognitive function. Interestingly, interleukin 6 (IL-6) infusion in young animals significantly reduced the production of AlloP compared with controls. It is notable that inhibition of IL-6 with its natural inhibitor, soluble membrane glycoprotein 130, significantly improved spatial memory in aged mice. These findings were supported by in vitro experiments in primary murine astrocyte cultures, indicating that IL-6 decreases production of AlloP and increases CORT levels. Our results indicate that age-related increases in IL-6 levels reduce progesterone substrate availability, resulting in a decline in AlloP levels and an increase in CORT. Furthermore, our results indicate that AlloP is a critical link between inflammatory cytokines and the age-related decline in cognitive function.


Subject(s)
Aging/physiology , Brain/metabolism , Cognition , Interleukin-6/metabolism , Pregnanolone/biosynthesis , Aging/metabolism , Animals , Brain/physiology , Male , Mice
3.
J Pharmacol Sci ; 143(3): 234-237, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32249061

ABSTRACT

We investigated whether benzothiazepines could produce anxiolytic effects via allopregnanolone (ALLO) biosynthesis in mice. We compared the behavioral effects caused by benzothiazepines to those caused by carbamazepine and sodium valproate. We found that a pretreatment with finasteride (a 5 alpha-reductase inhibitor) suppressed carbamazepine-induced anxiolytic effects but not the effects of sodium valproate. Similar to carbamazepine, diltiazem and JTV-519 displayed anxiolytic effects that were suppressed by a pretreatment with finasteride. We clearly demonstrate that the benzothiazepines, diltiazem and JTV-519, exert an anxiolytic-like effect via ALLO biosynthesis in mice.


Subject(s)
Anti-Anxiety Agents , Behavior, Animal/drug effects , Diltiazem/pharmacology , Pregnanolone/biosynthesis , Thiazepines/pharmacology , 5-alpha Reductase Inhibitors/pharmacology , Animals , Carbamazepine/antagonists & inhibitors , Carbamazepine/pharmacology , Diltiazem/antagonists & inhibitors , Dose-Response Relationship, Drug , Finasteride/pharmacology , Male , Mice, Inbred C57BL , Neurosteroids , Thiazepines/antagonists & inhibitors , Valproic Acid/pharmacology
4.
Biol Psychiatry ; 85(12): 1036-1045, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30955840

ABSTRACT

BACKGROUND: The endocannabinoid and neurosteroid systems regulate emotions and stress responses. Activation of peroxisome proliferator-activated receptor (PPAR)-α by the endocannabinoid congener N-palmitoylethanolamine (PEA) regulates pathophysiological systems (e.g., inflammation, oxidative stress) and induces peripheral biosynthesis of allopregnanolone, a gamma-aminobutyric acidergic neurosteroid implicated in mood disorders. However, effects of PPAR-α on emotional behavior are poorly understood. METHODS: We studied the impact of PPAR-α activation on emotional behavior in a mouse model of posttraumatic stress disorder. Neurosteroid levels before and after PEA treatment were measured by gas chromatography-mass spectrometry in relevant brain regions of socially isolated versus group-housed mice exposed to the contextual fear conditioning test, elevated plus maze test, forced swim test, and tail suspension test. Neurosteroidogenic enzyme levels were quantified in hippocampus by Western blot. RESULTS: PEA administered in a model of conditioned contextual fear reconsolidation blockade facilitated fear extinction and fear extinction retention and induced marked antidepressive- and anxiolytic-like effects in socially isolated mice with reduced brain allopregnanolone levels. These effects were mimicked by the PPAR-α synthetic agonists, fenofibrate and GW7647, and were prevented by PPAR-α deletion, PPAR-α antagonists, and neurosteroid-enzyme inhibitors. Behavioral improvements correlated with PEA-induced upregulation of PPAR-α, neurosteroidogenic enzyme expression, and normalization of corticolimbic allopregnanolone levels. CONCLUSIONS: This evidence supports a previously unknown role for PPAR-α in behavior regulation and suggests new strategies for the treatment of neuropsychopathologies characterized by deficient neurosteroidogenesis, including posttraumatic stress disorder and major depressive disorder.


Subject(s)
Emotions/physiology , Ethanolamines/administration & dosage , PPAR alpha/physiology , Palmitic Acids/administration & dosage , Pregnanolone/biosynthesis , Stress Disorders, Post-Traumatic/physiopathology , Amides , Animals , Anxiety/physiopathology , Disease Models, Animal , Emotions/drug effects , Fear/drug effects , Fear/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , PPAR alpha/metabolism , Social Isolation
5.
PLoS One ; 14(1): e0211266, 2019.
Article in English | MEDLINE | ID: mdl-30703109

ABSTRACT

Dysfunctions in the GABAergic system are associated with the pathogenesis of autism spectrum disorder (ASD). However, the mechanisms by which GABAergic system dysfunctions induce the pathophysiology of ASD remain unclear. We previously demonstrated that a selective type I 5α-reductase inhibitor SKF105111 (SKF) induced ASD-like behaviors, such as impaired sociability-related performance and repetitive grooming behaviors, in male mice. Moreover, the effects of SKF were caused by a decrease in the endogenous levels of allopregnanolone (ALLO), a positive allosteric modulator of the GABAA receptor. In this study, we used SKF-treated male mice as a putative animal model of ASD and examined the effects of Kami-shoyo-san (KSS) as an experimental therapeutic strategy for ASD. KSS is a traditional Kampo formula consisting of 10 different crude drugs and has been used for the treatment of neuropsychiatric symptoms. KSS dose-dependently attenuated sociability deficits and suppressed an increase in grooming behaviors in SKF-treated mice without affecting ALLO content in the prefrontal cortex. The systemic administration of the dopamine D1 receptor antagonist SCH23390 reversed the ameliorative effects of KSS. On the other hand, the dopamine D2 receptor antagonist sulpiride and GABAA receptor antagonist bicuculline only attenuated the ameliorative effect of KSS on repetitive self-grooming behaviors. The present results indicate that KSS improves SKF-induced ASD-like behaviors by facilitating dopamine receptor-mediated mechanisms and partly by neurosteroid-independent GABAA receptor-mediated neurotransmission. Therefore, KSS is a potential candidate for the treatment of ASD.


Subject(s)
Androstanes/adverse effects , Autism Spectrum Disorder/drug therapy , Drugs, Chinese Herbal/administration & dosage , Pregnanolone/biosynthesis , Animals , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/metabolism , Behavior, Animal/drug effects , Benzazepines/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/pharmacology , Grooming/drug effects , Humans , Male , Mice , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, GABA-A/metabolism , Treatment Outcome
6.
Biochem Pharmacol ; 161: 136-148, 2019 03.
Article in English | MEDLINE | ID: mdl-30668937

ABSTRACT

Gelsemine, the principal active alkaloid from Gelsemium sempervirens Ait., and koumine, the most dominant alkaloids from Gelsemium elegans Benth., produced antinociception in a variety of rodent models of painful hypersensitivity. The present study explored the molecular mechanisms underlying gelsemine- and koumine-induced mechanical antiallodynia in neuropathic pain. The radioligand binding and displacement assays indicated that gelsemine and koumine, like glycine, were reversible and orthosteric agonists of glycine receptors with full efficacy and probably acted on same binding site as the glycine receptor antagonist strychnine. Treatment with gelsemine, koumine and glycine in primary cultures of spinal neurons (but not microglia or astrocytes) concentration dependently increased 3α-hydroxysteroid oxidoreductase (3α-HSOR) mRNA expression, which was inhibited by pretreatment with strychnine but not the glial inhibitor minocycline. Intrathecal injection of gelsemine, koumine and glycine stimulated 3α-HSOR mRNA expression in the spinal cords of neuropathic rats and produced mechanical antiallodynia. Their spinal mechanical antiallodynia was completely blocked by strychnine, the selective 3α-HSOR inhibitor medroxyprogesterone acetate (MPA), 3α-HSOR gene silencer siRNA/3α-HSOR and specific GABAA receptor antagonist isoallopregnanolone, but not minocycline. All the results taken together uncovered that gelsemine and koumine are orthosteric agonists of glycine receptors, and produce mechanical antiallodynia through neuronal glycine receptor/3α-HSOR/allopregnanolone/GABAA receptor pathway.


Subject(s)
Alkaloids/metabolism , Gelsemium/metabolism , Hyperalgesia/metabolism , Indole Alkaloids/metabolism , Pregnanolone/biosynthesis , Receptors, Glycine/metabolism , Spinal Cord/metabolism , Alkaloids/therapeutic use , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Female , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Indole Alkaloids/therapeutic use , Male , Rats , Rats, Wistar , Spinal Cord/drug effects
7.
Cell Mol Neurobiol ; 39(4): 523-537, 2019 May.
Article in English | MEDLINE | ID: mdl-30187261

ABSTRACT

In the last decades, an active and stimulating area of research has been devoted to explore the role of neuroactive steroids in pain modulation. Despite challenges, these studies have clearly contributed to unravel the multiple and complex actions and potential mechanisms underlying steroid effects in several experimental conditions that mimic human chronic pain states. Based on the available data, this review focuses mainly on progesterone and its reduced derivative allopregnanolone (also called 3α,5α-tetrahydroprogesterone) which have been shown to prevent or even reverse the complex maladaptive changes and pain behaviors that arise in the nervous system after injury or disease. Because the characterization of new related molecules with improved specificity and enhanced pharmacological profiles may represent a crucial step to develop more efficient steroid-based therapies, we have also discussed the potential of novel synthetic analogs of allopregnanolone as valuable molecules for the treatment of neuropathic pain.


Subject(s)
Neuralgia/metabolism , Pregnanolone/metabolism , Progesterone/metabolism , Translational Research, Biomedical , Animals , Humans , Models, Biological , Neuralgia/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Pregnanolone/biosynthesis , Progesterone/biosynthesis , Progesterone/chemistry
8.
Pharmacol Ther ; 191: 190-206, 2018 11.
Article in English | MEDLINE | ID: mdl-29953900

ABSTRACT

Neurosteroids are neuroactive brain-born steroids. They can act through non-genomic and/or through genomic pathways. Genomic pathways are largely described for steroid hormones: the binding to nuclear receptors leads to transcription regulation. Pregnenolone, Dehydroepiandrosterone, their respective sulfate esters and Allopregnanolone have no corresponding nuclear receptor identified so far whereas some of their non-genomic targets have been identified. Neuroplasticity is the capacity that neuronal networks have to change their structure and function in response to biological and/or environmental signals; it is regulated by several mechanisms, including those that involve neurosteroids. In this review, after a description of their biosynthesis, the effects of Pregnenolone, Dehydroepiandrosterone, their respective sulfate esters and Allopregnanolone on their targets will be exposed. We then shall highlight that neurosteroids, by acting on these targets, can regulate neurogenesis, structural and functional plasticity. Finally, we will discuss the therapeutic potential of neurosteroids in the pathophysiology of neurological diseases in which alterations of neuroplasticity are associated with changes in neurosteroid levels.


Subject(s)
Nervous System Diseases/physiopathology , Neuronal Plasticity/physiology , Neurotransmitter Agents/metabolism , Animals , Dehydroepiandrosterone/biosynthesis , Dehydroepiandrosterone/metabolism , Humans , Nervous System Diseases/therapy , Neurogenesis/physiology , Neurotransmitter Agents/biosynthesis , Pregnanolone/biosynthesis , Pregnanolone/metabolism , Pregnenolone/biosynthesis , Pregnenolone/metabolism
9.
Biochem J ; 474(23): 3985-3999, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29074640

ABSTRACT

The 18 kDa translocator protein (TSPO) is a ubiquitous conserved outer mitochondrial membrane protein implicated in numerous cell and tissue functions, including steroid hormone biosynthesis, respiration, cell proliferation, and apoptosis. TSPO binds with high affinity to cholesterol and numerous compounds, is expressed at high levels in steroid-synthesizing tissues, and mediates cholesterol import into mitochondria, which is the rate-limiting step in steroid formation. In humans, the rs6971 polymorphism on the TSPO gene leads to an amino acid substitution in the fifth transmembrane loop of the protein, which is where the cholesterol-binding domain of TSPO is located, and this polymorphism has been associated with anxiety-related disorders. However, recent knockout mouse models have provided inconsistent conclusions of whether TSPO is directly involved in steroid synthesis. In this report, we show that TSPO deletion mutations in rat and its corresponding rs6971 polymorphism in humans alter adrenocorticotropic hormone-induced plasma corticosteroid concentrations. Rat tissues examined show increased cholesteryl ester accumulation, and neurosteroid formation was undetectable in homozygous rats. These results also support a role for TSPO ligands in diseases with steroid-dependent stress and anxiety elements.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Carrier Proteins/genetics , Hydrocortisone/blood , Polymorphism, Single Nucleotide , Receptors, GABA-A/genetics , Receptors, GABA/genetics , Adolescent , Adult , Animals , Base Sequence , Carrier Proteins/metabolism , Cholesterol Esters/biosynthesis , Cholesterol Esters/blood , Chorionic Gonadotropin/pharmacology , Cloning, Molecular , Corticosterone/biosynthesis , Corticosterone/blood , Embryo, Mammalian , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Hydrocortisone/biosynthesis , Male , Plasmids/chemistry , Plasmids/metabolism , Pregnanolone/biosynthesis , Pregnanolone/blood , Rats , Rats, Transgenic , Receptors, GABA/metabolism , Receptors, GABA-A/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Testosterone/biosynthesis , Testosterone/blood , Zinc Fingers , Zygote/drug effects , Zygote/growth & development , Zygote/metabolism
10.
Horm Behav ; 94: 106-113, 2017 08.
Article in English | MEDLINE | ID: mdl-28666923

ABSTRACT

Allopregnanolone, a neurosteroid whose levels rise throughout gestation, putatively stabilizes antenatal mood. The present study aimed to investigate associations of plasma allopregnanolone to antenatal depressive symptoms, as well as to genetic and obstetric factors. Allopregnanolone plasma levels from 284 pregnant women were measured around gestational week 18. Haplotype tag single nucleotide polymorphisms in the aldo-keto reductase family 1, members C2 and C4 (AKR1C2, AKR1C4), and steroid 5 alpha-reductase 1 and 2 (SRD5A1, and SRD5A2) genes were genotyped in a larger sample of pregnant women (n=1351). The Edinburgh Postnatal Depression Scale (EPDS) was administered via web-questionnaires in gestational weeks 17 and 32. Demographic and obstetric data was retrieved from web-questionnaires and medical records. There was no association between allopregnanolone levels and depressive symptoms. Furthermore, no associations between allopregnanolone level and synthesis pathway genotypes were found after accounting for multiple comparisons. However, exploratory analyses suggested that the women who were homozygous for the minor allele of the AKR1C2 polymorphism rs1937863 had nominally lower allopregnanolone levels and lower depression scores in gestational week 17, but also the highest increase in depression scores between week 17 and 32. Additionally, higher body mass index was associated with lower allopregnanolone levels. The results do not support second trimester plasma allopregnanolone as a mood stabilizing factor. However, we speculate that AKR1C2 variation may alter the susceptibility to depressive symptoms through effects on central allopregnanolone synthesis. Another implication of this study is that the relationship between neuroactive steroids and obesity in pregnancy deserves to be investigated.


Subject(s)
Depression/genetics , Metabolic Networks and Pathways/genetics , Polymorphism, Single Nucleotide , Pregnancy Complications/genetics , Pregnanolone/biosynthesis , Pregnanolone/blood , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Adult , Depression/blood , Depression/metabolism , Depression, Postpartum/blood , Depression, Postpartum/genetics , Depression, Postpartum/metabolism , Female , Genetic Association Studies , Humans , Hydroxysteroid Dehydrogenases/genetics , Membrane Proteins/genetics , Oxidoreductases/genetics , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/metabolism , Psychiatric Status Rating Scales
11.
Synapse ; 71(8)2017 08.
Article in English | MEDLINE | ID: mdl-28407365

ABSTRACT

The neurosteroid allopregnanolone (3α, 5α-tetra-hydroprogesterone: ALLO) elicits anxiolytic, anticonvulsant, and hypnotic anesthetic effects in vivo similar to those induced by other positive allosteric modulators of the GABAA receptor. Endogenous ALLO has been shown to be rapidly elevated in the brain by acute stress paradigms, such as immobilization, in animal models. The present study was designed to ascertain the role of neurosteroid biosynthesis in the anxiety-like behavior induced by immobilization stress. Mice were exposed to an immobilization stressor for 2 h. After 24 h, the mice that had been immobilized did not behave significantly differently in the elevated plus maze (EPM) test and in the elevated open platform (EOP) test than the mice that had not been immobilized. In contrast, finasteride-pretreated immobilization stressed mice did behave significantly differently in the EPM and EOP tests. These findings suggest that ALLO biosynthesis contributes to stress resistance. Furthermore, the ALLO mimetic drug alfaxalone appeared to antagonize the effects of finasteride by significantly changing the behavior in the EPM test or in the EOP test in finasteride (10 mg kg-1 )-pretreated immobilized mice. In addition, alfaxalone, unlike diazepam, did not affect the muscle tone of the mice, as measured by the grip strength test. These results suggest that alfaxalone is a promising anxiolytic candidate lacking benzodiazepine-like muscle-relaxant effects.


Subject(s)
Anxiety Disorders/metabolism , Pregnanolone/biosynthesis , Stress, Psychological/metabolism , 5-alpha Reductase Inhibitors/pharmacology , Animals , Anxiety Disorders/drug therapy , Diazepam/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Finasteride/pharmacology , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Male , Mice, Inbred C57BL , Muscle Relaxants, Central/pharmacology , Muscle Strength/drug effects , Muscle Strength/physiology , Muscle Tonus/drug effects , Muscle Tonus/physiology , Pregnanediones/pharmacology , Restraint, Physical , Stress, Psychological/drug therapy
12.
J Ethnopharmacol ; 198: 324-330, 2017 Feb 23.
Article in English | MEDLINE | ID: mdl-27993636

ABSTRACT

ETHNOPHARMACOLOGY RELEVANCE: Post-traumatic stress disorder (PTSD) is a severe psychiatric disorder that is characterized by symptoms of re-experiencing, avoidance and hyperarousal, as well as social and professional dysfunction at least one month after the exposure to a traumatic event. Biosynthesis of allopregnanolone has been suggested as one of the important contributors to PTSD. Albiflorin (AF) extracted from Radix paeoniae Alba had been shown to be effective in the therapy of depression. However, few studies were concerned about the anti-PTSD-like effects of AF. AIM OF THE STUDY: The current study aimed to evaluate the anti-PTSD-like effects of AF in an animal model and its possible mechanism. MATERIALS AND METHODS: To evaluate this, the single prolonged stress (SPS) model was used in the present study. The SPS rats were administered by AF (at doses of 3.5, 7 and 14.0mg/kg, i.g.) after induction of SPS from days 2-13. After the exposure to SPS, behavioral assessments were conducted, including contextual fear paradigm (CFP), elevated plus-maze test (EPMT), open-field test (OFT). The rats were decapitated at the end of the behavioral tests and levels of allopregnanolone in prefrontal cortex, hippocampus and amygdala were measured by enzyme linked immunosorbent assay (ELISA). RESULTS: It had been shown that behavioral deficits of SPS rats were reversed by AF (7.0 and 14.0mg/kg, i.g.), which attenuated the PTSD-like associated contextual freezing behavior in CFP and improved PTSD-like associated anxiogenic behavior in EPMT without affecting locomotor activity in OFT. Moreover, decreased levels of allopregnanolone in prefrontal cortex, hippocampus, and amygdala were reversed by AF (7.0 and 14.0mg/kg, i.g.), respectively. CONCLUSION: In summary, the present study indicated that AF exerted the anti-PTSD-like effects, which maybe associated with allopregnanolone biosynthesis in the brain.


Subject(s)
Bridged-Ring Compounds/pharmacology , Paeonia/chemistry , Stress Disorders, Post-Traumatic/drug therapy , Amygdala/drug effects , Animals , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/isolation & purification , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fear/drug effects , Hippocampus/drug effects , Male , Maze Learning/drug effects , Prefrontal Cortex/drug effects , Pregnanolone/biosynthesis , Rats , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/pathology
13.
Neuropharmacology ; 111: 142-159, 2016 12.
Article in English | MEDLINE | ID: mdl-27596950

ABSTRACT

We previously reported that elevated pressure induces axonal swelling and facilitates the synthesis of the neurosteroid, allopregnanolone (AlloP), in the ex vivo rat retina. Exogenously applied AlloP attenuates the axonal swelling, suggesting that the neurosteroid plays a neuroprotective role against glaucomatous pressure-induced injuries, although mechanisms underlying neurosteroidogenesis have not been clarified. The aim of this study was to determine whether AlloP synthesis involves activation of translocator protein 18 kD (TSPO) and whether TSPO modulates pressure-induced retinal injury. Ex vivo rat retinas were exposed to various pressures (10, 35, or 75 mmHg) for 24 h. Expression of TSPO, 5α-reductase (5aRD), and AlloP was examined by quantitative real-time RT-PCR, ELISA, immunohistochemistry, and LC-MS/MS. We also examined the effects of TSPO ligands on AlloP synthesis and retinal damage. In this acute model, quantitative real-time RT-PCR and ELISA analyses revealed that elevated pressure facilitated TSPO expression. Similarly, these methods also detected enhanced 5aRD (mostly type II), which was observed in retinal ganglion cells (RGC) and the inner nuclear layer (INL). Atriol, a TSPO antagonist, suppressed pressure mediated AlloP synthesis and induced more severe histological changes in the inner retina when combined with elevated pressure. PK11195, a TSPO ligand that facilitates AlloP synthesis by itself, remarkably diminished pressure-mediated retinal degeneration. These results suggest that AlloP synthesis is induced by sequential activation of TSPO and 5aRD in an ex vivo glaucoma model, and that TSPO agonists may serve as potential therapeutic agents for the prevention of pressure-induced retinal damage.


Subject(s)
Carrier Proteins/metabolism , Cholestenone 5 alpha-Reductase/metabolism , Glaucoma/metabolism , Pregnanolone/biosynthesis , Receptors, GABA-A/metabolism , 5-alpha Reductase Inhibitors/administration & dosage , Animals , Disease Models, Animal , Dutasteride/administration & dosage , Glaucoma/pathology , In Vitro Techniques , Isoquinolines/administration & dosage , Male , Pressure , Rats , Rats, Sprague-Dawley , Retina/pathology , Retina/ultrastructure , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Up-Regulation
14.
Cancer Biomark ; 17(1): 11-6, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27062569

ABSTRACT

Breast cancer is currently the most common malignancy affecting women worldwide. It had been shown that the allopregnanolone biosynthesis was associated with tumorigenesis and PK11195, the Translocator Protein 18 KDa (TSPO) antagonist, had the effects of the allopregnanolone biosynthesis. However, little is known about the association between the effects of PK11195 on the breast cancer and the allopregnanolone biosynthesis. To evaluate this, the breast cancer cell lines MCF-7 and T47D were cultured. Cell viability and proliferation were determined by CCK-8 assay. The IC50 of PK11195 on the MCF-7 and T47D were 5.4 nM and 6 nM. The cell viability and proliferation of AC-5216 (TSPO selective ligand, 3 and 6 nM) was blocked by PK11195 (5.4 nM and 6 nM). Moreover, we evaluated the role of allopregnanolone biosynthesis in the effects of TSPO on breast cancer. Enzyme-Linked ImmunoSorbent Assay (ELISA) was used in the measurement of the allopregnanolone level. We found that the allopregnanolone level was increased by AC-5216 (3 and 6 nM) and the increase was reversed by PK11195 (5.4 nM and 6 nM, resepectively) in MCF-7 and T47D. The TSPO mRNA level was determined by real time polymerase chain reaction (PCR). The TSPO mRNA level were increased by AC-5216 (6 nM), which the increases were reversed by PK11195 (5.4 nM and 6 nM, resepectively) in MCF-7 and T47D. Collectedly, it firstly indicated that the effects of PK11195 on MCF-7 and T47D were associated with the decrease of allopregnanolone biosynthesis, which was mediated by TSPO.


Subject(s)
Antineoplastic Agents/pharmacology , Isoquinolines/pharmacology , Pregnanolone/biosynthesis , Receptors, GABA/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Gene Expression , Humans , MCF-7 Cells , RNA, Messenger/genetics , Receptors, GABA/genetics
15.
Cancer Biomark ; 16(1): 65-9, 2016.
Article in English | MEDLINE | ID: mdl-26835706

ABSTRACT

Meningioma is one of the common brain tumors in adults. It had been shown that the allopregnanolone biosynthesis was associated with tumorigenesis and PK11195, the translocator protein 18 KDa (TSPO) antagonist, had the effects of the allopregnanolone biosynthesis. However, little is known about the association between the effects of PK11195 on meningioma and the allopregnanolone biosynthesis. To evaluate this, the meningioma cell line IOMM-LEE was applied. Cell viability and proliferation were determined by CCK-8 assay. The IC50 of PK11195 on the IOMM-LEE was 1.505 ± 0.08 nM. The cell viability and proliferation of AC-5216 (TSPO selective ligand, 2 and 4 nM) was blocked by PK11195 (1.5 nM). Further, we evaluated the role of allopregnanolone biosynthesis in the effects of TSPO on meningioma. Enzyme-Linked ImmunoSorbent Assay (ELISA) was used in the measurement of the allopregnanolone level. It showed that the allopregnanolone level was increased by AC-5216 (2 and 4 nM) and the increase was reversed by PK11195 (1.5 nM). Collectedly, it firstly indicated that the effects of PK11195 on meningioma were relevant to the decrease of allopregnanolone biosynthesis, which was mediated by TSPO.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Drug Resistance, Neoplasm , Isoquinolines/pharmacology , Meningioma/metabolism , Pregnanolone/biosynthesis , Receptors, GABA/metabolism , Cell Line, Tumor , Humans
16.
Neurosci Lett ; 602: 162-6, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26160034

ABSTRACT

Post-traumatic stress disorder (PTSD) is a severe psychiatric condition defined as a "trauma and stress-related disorder". Dampened allopregnanolone biosynthesis has been implicated as a possible contributor to PTSD aetiology. Free and Easy Wanderer Plus (FEWP) is a traditional Chinese medicine previously shown to be effective in PTSD treatment. However, little is known about the role of allopregnanolone in the anti-PTSD effects of FEWP. To evaluate this, the single prolonged stress (SPS) model was used in the present study. SPS-induced rats were administered FEWP (at doses of 2.5, 5.0 and 10.0 mg/kg, p.o.) after induction of SPS from days 2 through 15. After exposure to SPS, behavioral assessments were determined, including the open-field test, the contextual fear paradigm, and the elevated plus-maze test. The experimental model rats were decapitated at the end of the behavioral tests and the level of allopregnanolone in the prefrontal cortex, hippocampus and amygdala was measured by enzyme linked immunosorbent assay (ELISA). The behavioral deficits of the SPS-induced rats were significantly reversed by FEWP (at doses of 5.0 and 10.0 mg/kg, p.o.). The level of allopregnanolone was increased by administration of FEWP. In summary, this study indicated that the anti-PTSD effects of FEWP were associated with allopregnanolone biosynthesis.


Subject(s)
Behavior, Animal/drug effects , Drugs, Chinese Herbal/therapeutic use , Pregnanolone/biosynthesis , Stress Disorders, Post-Traumatic/drug therapy , Animals , Fear , Male , Maze Learning/drug effects , Motor Activity/drug effects , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/psychology
17.
Neurosci Lett ; 595: 94-8, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25800108

ABSTRACT

Anxiety disorders are the serious and burdensome psychiatric illnesses, which are closely correlated with allopregnanolone. The down-regulation of allopregnanolone biosynthesis has been implicated as the possible contributor to the aetiology of anxiety disorders. Free and easy wanderer plus (FEWP) is a well-known traditional Chinese medicine that had been shown to be effective in various mood disorders. The purpose of the present study was to evaluate the anxiolytic-like effect of FEWP and its association with the level of allopregnanolone in the brain. The animal behavioral tests were processed by the acute FEWP (2.5, 5 and 10mg/kg, p.o.) treatment. It had been shown that FEWP produced anxiolytic-like effects in behavioral models, including novelty suppressed feeding (5, 10mg/kg, p.o.), Vogel-type conflict test (10mg/kg, p.o.), elevated plus-maze test (5, 10mg/kg, p.o.). The animals were decapitated after the end of the behavioral tests and measured the allopregnanolone level of the prefrontal cortex and hippocampus by enzyme-linked immunosorbent assay (ELISA). The allopregnanolone level of the prefrontal cortex and hippocampus was increased by administration of FEWP (5, 10mg/kg, p.o.). Overall, these results indicated that FEWP exerts anxiolytic-like effects that were associated with the stiumlation of the allopregnanolone biosynthesis.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Drugs, Chinese Herbal/therapeutic use , Pregnanolone/biosynthesis , Animals , Anxiety/psychology , Conflict, Psychological , Feeding Behavior/drug effects , Male , Maze Learning/drug effects , Mice, Inbred ICR , Rats, Sprague-Dawley , Reaction Time/drug effects
18.
PLoS One ; 10(3): e0120473, 2015.
Article in English | MEDLINE | ID: mdl-25785994

ABSTRACT

Neurosteroids can modulate the activity of the GABAA receptors, and thus affect anxiety-like behaviors. The non-benzodiazepine anxiolytic compound etifoxine has been shown to increase neurosteroid concentrations in brain tissue but the mode of action of etifoxine on neurosteroid formation has not yet been elucidated. In the present study, we have thus investigated the effect and the mechanism of action of etifoxine on neurosteroid biosynthesis using the frog hypothalamus as an experimental model. Exposure of frog hypothalamic explants to graded concentrations of etifoxine produced a dose-dependent increase in the biosynthesis of 17-hydroxypregnenolone, dehydroepiandrosterone, progesterone and tetrahydroprogesterone, associated with a decrease in the production of dihydroprogesterone. Time-course experiments revealed that a 15-min incubation of hypothalamic explants with etifoxine was sufficient to induce a robust increase in neurosteroid synthesis, suggesting that etifoxine activates steroidogenic enzymes at a post-translational level. Etifoxine-evoked neurosteroid biosynthesis was not affected by the central-type benzodiazepine (CBR) receptor antagonist flumazenil, the translocator protein (TSPO) antagonist PK11195 or the GABAA receptor antagonist bicuculline. In addition, the stimulatory effects of etifoxine and the triakontatetraneuropeptide TTN, a TSPO agonist, were additive, indicating that these two compounds act through distinct mechanisms. Etifoxine also induced a rapid stimulation of neurosteroid biosynthesis from frog hypothalamus homogenates, a preparation in which membrane receptor signalling is disrupted. In conclusion, the present study demonstrates that etifoxine stimulates neurosteroid production through a membrane receptor-independent mechanism.


Subject(s)
17-alpha-Hydroxypregnenolone/agonists , Anti-Anxiety Agents/pharmacology , Dehydroepiandrosterone/agonists , Hypothalamus/drug effects , Oxazines/pharmacology , Pregnanolone/agonists , Progesterone/agonists , 17-alpha-Hydroxypregnenolone/metabolism , 20-alpha-Dihydroprogesterone/antagonists & inhibitors , 20-alpha-Dihydroprogesterone/biosynthesis , Animals , Bicuculline/pharmacology , Complex Mixtures/chemistry , Dehydroepiandrosterone/biosynthesis , Dose-Response Relationship, Drug , Flumazenil/pharmacology , GABA Modulators/pharmacology , GABA-A Receptor Antagonists/pharmacology , Gene Expression , Hypothalamus/metabolism , Isoquinolines/pharmacology , Male , Neuropeptides/pharmacology , Peptide Fragments/pharmacology , Pregnanolone/biosynthesis , Progesterone/biosynthesis , Rana esculenta , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Tissue Culture Techniques
19.
Br J Pharmacol ; 171(24): 5870-80, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25161074

ABSTRACT

BACKGROUND AND PURPOSE: Fluoxetine, a selective serotonin reuptake inhibitor, elevates brain concentrations of the neuroactive progesterone metabolite allopregnanolone, an effect suggested to underlie its use in the treatment of premenstrual dysphoria. One report showed fluoxetine to activate the aldo-keto reductase (AKR) component of 3α-hydroxysteroid dehydrogenase (3α-HSD), which catalyses production of allopregnanolone from 5α-dihydroprogesterone. However, this action was not observed by others. The present study sought to clarify the site of action for fluoxetine in elevating brain allopregnanolone. EXPERIMENTAL APPROACH: Adult male rats and female rats in dioestrus were treated with fluoxetine and their brains assayed for allopregnanolone and its precursors, progesterone and 5α-dihydroprogesterone. Subcellular fractions of rat brain were also used to investigate the actions of fluoxetine on 3α-HSD activity in both the reductive direction, producing allopregnanolone from 5α-dihydroprogesterone, and the reverse oxidative direction. Fluoxetine was also tested on these recombinant enzyme activities expressed in HEK cells. KEY RESULTS: Short-term treatment with fluoxetine increased brain allopregnanolone concentrations in female, but not male, rats. Enzyme assays on native rat brain fractions and on activities expressed in HEK cells showed fluoxetine did not affect the AKR producing allopregnanolone from 5α-dihydroprogesterone but did inhibit the microsomal dehydrogenase oxidizing allopregnanolone to 5α-dihydroprogesterone. CONCLUSIONS AND IMPLICATIONS: Fluoxetine elevated allopregnanolone in female rat brain by inhibiting its oxidation to 5α-dihydroprogesterone by a microsomal dehydrogenase. This is a novel site of action for fluoxetine, with implications for the development of new agents and/or dosing regimens to raise brain allopregnanolone.


Subject(s)
3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/antagonists & inhibitors , Brain/drug effects , Fluoxetine/pharmacology , Pregnanolone/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/metabolism , 5-alpha-Dihydroprogesterone/metabolism , Aldehyde Reductase/drug effects , Aldehyde Reductase/metabolism , Aldo-Keto Reductases , Animals , Brain/metabolism , Female , HEK293 Cells , Humans , Male , Pregnanolone/biosynthesis , Progesterone/metabolism , Rats
20.
J Neurosci ; 33(42): 16617-26, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24133265

ABSTRACT

Hypothalamospinal control of spinal pain processing by oxytocin (OT) has received a lot of attention in recent years because of its potency to reduce pain symptoms in inflammatory and neuropathic conditions. However, cellular and molecular mechanisms underlying OT spinal antinociception are still poorly understood. In this study, we used biochemical, electrophysiological, and behavioral approaches to demonstrate that OT levels are elevated in the spinal cord of rats exhibiting pain symptoms, 24 h after the induction of inflammation with an intraplantar injection of λ-carrageenan. Using a selective OT receptor antagonist, we demonstrate that this elevated OT content is responsible for a tonic analgesia exerted on both mechanical and thermal modalities. This phenomenon appeared to be mediated by an OT receptor-mediated stimulation of neurosteroidogenesis, which leads to an increase in GABA(A) receptor-mediated synaptic inhibition in lamina II spinal cord neurons. We also provide evidence that this novel mechanism of OT-mediated spinal antinociception may be controlled by extracellular signal-related protein kinases, ERK1/2, after OT receptor activation. The oxytocinergic inhibitory control of spinal pain processing is emerging as an interesting target for future therapies since it recruits several molecular mechanisms, which are likely to exert a long-lasting analgesia through nongenomic and possibly genomic effects.


Subject(s)
Analgesia , Inhibitory Postsynaptic Potentials/physiology , Oxytocin/metabolism , Pain/metabolism , Pregnanolone/biosynthesis , Receptors, GABA-A/metabolism , Spinal Cord/metabolism , Animals , Carrageenan , Hormone Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Male , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Neurons/drug effects , Neurons/metabolism , Oxytocin/analogs & derivatives , Oxytocin/pharmacology , Pain/chemically induced , Rats , Rats, Sprague-Dawley , Receptors, Oxytocin/antagonists & inhibitors , Spinal Cord/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL