Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 11(11): e10378, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31566924

ABSTRACT

Current treatments in multiple sclerosis (MS) are modulating the inflammatory component of the disease, but no drugs are currently available to repair lesions. Our study identifies in MS patients the overexpression of Plexin-A1, the signalling receptor of the oligodendrocyte inhibitor Semaphorin 3A. Using a novel type of peptidic antagonist, we showed the possibility to counteract the Sema3A inhibitory effect on oligodendrocyte migration and differentiation in vitro when antagonizing Plexin-A1. The use of this compound in vivo demonstrated a myelin protective effect as shown with DTI-MRI and confirmed at the histological level in the mouse cuprizone model of induced demyelination/remyelination. This effect correlated with locomotor performances fully preserved in chronically treated animals. The administration of the peptide also showed protective effects, leading to a reduced severity of demyelination in the context of experimental autoimmune encephalitis (EAE). Hence, the disruption of the inhibitory microenvironmental molecular barriers allows normal myelinating cells to exert their spontaneous remyelinating capacity. This opens unprecedented therapeutic opportunity for patients suffering a disease for which no curative options are yet available.


Subject(s)
Multiple Sclerosis/physiopathology , Nerve Tissue Proteins/metabolism , Oligodendroglia/physiology , Receptors, Cell Surface/metabolism , Remyelination , Semaphorin-3A/metabolism , Signal Transduction , Animals , Brain/diagnostic imaging , Cell Line , Cell Movement , Cell Proliferation , Disease Models, Animal , Magnetic Resonance Imaging , Mice, Inbred C57BL , Nerve Tissue Proteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors
2.
Alzheimers Res Ther ; 8: 37, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27601032

ABSTRACT

Gamma-hydroxybutyrate (GHB or Xyrem(R)) is frequently used in humans for several clinical indications, including anesthesia, narcolepsy/cataplexy, and alcohol-withdrawal symptoms. Pharmacological effects induced in the brain by therapeutic doses of Xyrem(R) are generally GABAergic-dependent. These effects allow sedation, stress/anxiety reduction, deep sleep induction, decrease of neuroinflammation, and neuroprotection. Furthermore, Xyrem(R) promotes the expression of pivotal genes reducing toxic proteinopathies, as demonstrated in laboratory animal models. Altogether, these data represent additional evidence to suggest that Xyrem(R) may be tested during repeated short periods in populations at risk for Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/administration & dosage , Sodium Oxybate/administration & dosage , Animals , Humans
3.
Sci Rep ; 6: 22542, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26935652

ABSTRACT

Patients with schizophrenia have often been described as insensitive to nociceptive signals, but objective evidence is sparse. We address this question by combining subjective behavioral and objective neurochemical and neurophysiological measures. The present study involved 21 stabilized and mildly symptomatic patients with schizophrenia and 21 control subjects. We applied electrical stimulations below the pain threshold and assessed sensations of pain and unpleasantness with rating scales, and Somatosensory Evoked Potentials (SEPs/EEG). We also measured attention, two neurochemical stress indices (ACTH/cortisol), and subjective VEPs/EEG responses to visual emotional stimuli. Our results revealed that, subjectively, patients' evaluations do not differ from controls. However, the amplitude of EEG evoked potentials was greater in patients than controls as early as 50 ms after electrical stimulations and beyond one second after visual processing of emotional pictures. Such responses could not be linked to the stress induced by the stimulations, since stress hormone levels were stable. Nor was there a difference between patients and controls in respect of attention performance and tactile sensitivity. Taken together, all indices measured in patients in our study were either heightened or equivalent relative to healthy volunteers.


Subject(s)
Electroencephalography , Emotions , Evoked Potentials, Somatosensory , Pain/physiopathology , Schizophrenia/physiopathology , Stress, Psychological/physiopathology , Adult , Electric Stimulation , Female , Humans , Male , Middle Aged
4.
Med Res Rev ; 36(3): 363-88, 2016 May.
Article in English | MEDLINE | ID: mdl-26739481

ABSTRACT

γ-Hydroxybutyrate (GHB) is both a natural brain compound with neuromodulatory properties at central GABAergic synapses (micromolar concentration range) and also a drug (Xyrem(R) ) clinically used for the treatment of various neurological symptoms (millimolar dose range). However, this drug has abuse potential and can be addictive for some patients. Here, we review the basic mechanistic role of endogenous GHB in brain as well as the properties and mechanisms of action for therapeutic clinical doses of exogenous GHB. Several hypotheses are discussed with a preference for a molecular mechanism that conciliates most of the findings available. This conciliatory model may help for the design of GHB-like drugs active at lower doses and devoid of major side effects.


Subject(s)
Brain/metabolism , Sodium Oxybate/metabolism , Humans
5.
Neurobiol Aging ; 36(2): 832-44, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25457559

ABSTRACT

The chronic decrease of brain amyloid-ß (Aß) peptides is an emerging therapeutic for Alzheimer's disease, but no such treatment has achieved clinical validation yet. In vivo, some brain proteases, including neprilysin, possess the ability of degrading Aß and experimental data suggest their exploitation in strategies to reduce cerebral Aß concentration. Previous studies have shown that pharmacologic doses of gamma-hydroxybutyrate (sodium oxybate or Xyrem) induce histone deacetylases (HDACs) inhibition and neprilysin gene expression. Here, we demonstrate that brain neprilysin overexpression induced in vivo by repeated gamma-hydroxybutyrate autoadministration reduces cerebral Aß contents and prevents cognitive deficits in APPSWE mice. Oral gamma-hydroxybutyrate also counteracted phosphoramidon-induced brain neprilysin inhibition and Aß accumulation. HDACs activities in SH-SY5Y cells were inhibited by gamma-hydroxybutyrate which did not affect amyloid peptide precursor intracellular domain. Together, our results suggest that gamma-hydroxybutyrate, acting via HDAC inhibition, upregulates neprilysin to reduce Aß level and related memory deficits. Because gamma-hydroxybutyrate doses used herein are clinically relevant, our data suggest that chronic oral administration of gamma-hydroxybutyrate or its analogs may be considered for strategies against presymptomatic or established Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Sodium Oxybate/administration & dosage , Sodium Oxybate/pharmacology , Administration, Oral , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cells, Cultured , Cognition/drug effects , Disease Models, Animal , Female , Gene Expression/drug effects , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Mice , Molecular Targeted Therapy , Neprilysin/genetics , Neprilysin/metabolism
6.
Behav Brain Res ; 253: 90-4, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23850351

ABSTRACT

The chemical compound gelsemine is the major active principle of the yellow jasmine (Gelsemium) that is generally claimed as possessing anxiolytic properties based on empirical and indirect knowledge. Surprisingly, gelsemine effect on anxiety has until now received only little attention. Here, we used the well-validated method for anxiety assessment, the elevated plus-maze combined with video-tracking, to measure gelsemine action on rat anxiety-like behavior. Rats were intraperitoneally injected (500µl/daily/7days) with gelsemine (10(-6), 10(-10) or 10(-14)M) or control solution. Diazepam (DZP) was used as positive standard anxiolytic and additional controls were naive rats similarly manipulated except being injected. Gelsemine or diazepam treatment did not affect the number of closed arm entries and rears illustrating the rat general activity. In contrast, gelsemine (10(-6) to 10(-10)M) or DZP increased dose-dependently the number of entries and the percent of time spent in the open arms indicating that gelsemine is an anxiolytic. Consistently, we observed that gelsemine (10(-6) to 10(-10)M) or DZP also decreased dose-dependently the percent of protected stretched attend postures, an ethological index of anxiety-like state. Altogether, our results constitute a solid set of fundamental data directly demonstrating anxiolytic properties of gelsemine. The report also opens new perspectives for the development of safe and effective gelsemine- or Gelsemium-based strategies against pathological anxiety.


Subject(s)
Alkaloids/pharmacology , Anti-Anxiety Agents , Anxiety/psychology , Animals , Behavior, Animal/drug effects , Diazepam/pharmacology , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Gelsemium/chemistry , Injections, Intraperitoneal , Male , Maze Learning/drug effects , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley
7.
Neuropharmacology ; 70: 254-60, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23422298

ABSTRACT

Kynurenic acid (KYNA), one of the main product of the kynurenine pathway originating from tryptophan, is considered to be neuroprotective. Dysregulation of KYNA activity is thought to be involved in neurodegenerative diseases, the physiopathology of which evokes excitotoxicity, oxidative stress and/or protein aggregation. The neuroprotective effect of KYNA is generally attributed to its antagonistic action on NMDA receptors. However, this single target action appears insufficient to support KYNA beneficial effects against complex neurodegenerative processes including neuroinflammation, ß-amyloid peptide (Aß) toxicity and apoptosis. Novel insights are therefore required to elucidate KYNA neuroprotective mechanisms. Here, we combined cellular, biochemical, molecular and pharmacological approaches to demonstrate that low micromolar concentrations of KYNA strongly induce neprilysin (NEP) gene expression, protein level and enzymatic activity increase in human neuroblastoma SH-SY5Y cells. Furthermore, our studies revealed that KYNA exerts a protective effect on SH-SY5Y cells by increasing their viability through a mechanism independent from NMDA receptors. Interestingly, KYNA also induced NEP activity and neuroprotection in mouse cortical neuron cultures the viability of which was more promoted than SH-SY5Y cell survival under KYNA treatment. KYNA-evoked neuroprotection disappeared in the presence of thiorphan, an inhibitor of NEP activity. NEP is a well characterized metallopeptidase whose deregulation leads to cerebral Aß accumulation and neuronal death in Alzheimer's disease. Therefore, our results suggest that a part of the neuroprotective role of KYNA may depend on its ability to induce the expression and/or activity of the amyloid-degrading enzyme NEP in nerve cells.


Subject(s)
Kynurenic Acid/pharmacology , Neprilysin/biosynthesis , Neurons/physiology , Neuroprotective Agents/pharmacology , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction , Gene Expression/drug effects , Humans , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/antagonists & inhibitors , Mice , Neprilysin/antagonists & inhibitors , Neprilysin/metabolism , Neuroprotective Agents/metabolism , Primary Cell Culture , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Thiorphan/pharmacology
8.
Pain ; 150(3): 522-534, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20605070

ABSTRACT

The enzyme 3alpha-hydroxysteroid oxido-reductase (3alpha-HSOR) catalyzes the synthesis and bioavailability of 3alpha,5alpha-neurosteroids as allopregnanolone (3alpha,5alpha-THP) which activates GABA(A) receptors and blocks T-type calcium channels involved in pain mechanisms. Here, we used a multidisciplinary approach to demonstrate that 3alpha-HSOR is a cellular target the modulation of which in dorsal root ganglia (DRG) may contribute to suppress pain resulting from peripheral nerve injury. Immunohistochemistry and confocal microscope analyses showed 3alpha-HSOR-immunostaining in naive rat DRG sensory neurons and glial cells. Pulse-chase, high performance liquid chromatography and Flo/One characterization of neurosteroids demonstrated 3alpha,5alpha-THP production in DRG. Behavioral methods allowed identification of pain symptoms (thermal and mechanical hyperalgesia and/or allodynia) in rats subjected to sciatic nerve chronic constriction injury (CCI). Reverse transcription and real-time polymerase chain reaction revealed that 3alpha-HSOR mRNA concentration in CCI-rat ipsilateral DRG, 5-fold higher than in contralateral DRG, was also 4- to 6-fold elevated than that in sham-operated or naive rat DRG. Consistently, Western blotting confirmed increased 3alpha-HSOR protein levels in CCI-rat ipsilateral DRG and double immunolabeling showed that 3alpha-HSOR overexpression occurred in DRG neurons but not in glia. Functional plasticity of 3alpha-HSOR leading to increased 3alpha,5alpha-THP production was evidenced in CCI-rat DRG. Interestingly, behavioral and molecular time-course investigations revealed that 3alpha-HSOR gene upregulation was correlated to pain symptom development. Most importantly, in vivo knockdown of 3alpha-HSOR expression in healthy rat DRG using 6-carboxyfluorescein-3alpha-HSOR-siRNA exacerbated thermal and mechanical pain perceptions. This paper is the first to show that siRNA-induced knockdown of a key neurosteroid-synthesizing enzyme directly affects an important function as nociception. Hopefully, these results may be useful for the development of novel analgesics.


Subject(s)
3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation, Enzymologic/physiology , Neurons/metabolism , Pain/pathology , Animals , Cell Nucleus/metabolism , Chromatography, High Pressure Liquid/methods , Cytoplasm/drug effects , Cytoplasm/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic/drug effects , Hyperalgesia/physiopathology , Male , Neurons/drug effects , Neurons/ultrastructure , Pain/drug therapy , Pain/etiology , Pain Perception/drug effects , Pain Threshold/physiology , Peripheral Nervous System Diseases/complications , Phosphopyruvate Hydratase/metabolism , Physical Stimulation/methods , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Steroids/metabolism , Time Factors , Tritium/metabolism
9.
Front Biosci (Elite Ed) ; 2(4): 1528-37, 2010 06 01.
Article in English | MEDLINE | ID: mdl-20515824

ABSTRACT

To characterize endogenous molecules regulating nociception, various groups have focused on dehydroepiandrosterone (DHEA). Indeed, DHEA modulates NMDA and P2X receptors which control neurobiological activities including nociception. Thus, various results were published on DHEA ability to regulate nociception but the data were interpreted separately. To provide an overview, we analyzed here the current knowledge on DHEA regulatory action on the spinal cord (SC) which is pivotal for nociception. DHEA endogenously synthesized in the SC appears as a key factor regulating nociception. However, DHEA effects on nociceptive mechanisms are complex. Acute DHEA treatment exerts a biphasic effect on nociception (a rapid pro-nociceptive action and a delayed anti-nociceptive effect). Chronic DHEA treatment increased basal nociceptive thresholds in neuropathic and control rats, suggesting that androgenic metabolites of DHEA exerted analgesic effects while DHEA itself caused a rapid pro-nociceptive action. To get more insights into DHEA effects on nociception, we provided a hypothetical scheme recapitulating cellular mechanisms of action of DHEA in the control of nociception. Perspective is opened for the development of DHEA-based strategies against pathological pain.


Subject(s)
Analgesia , Dehydroepiandrosterone/physiology , Spinal Cord/physiology , Animals , Dehydroepiandrosterone/biosynthesis , Humans , Spinal Cord/metabolism
10.
Physiol Genomics ; 41(2): 146-60, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20103696

ABSTRACT

γ-Hydroxybutyrate (GHB) is a natural brain neuromodulator that has its own enzymatic machinery for synthesis and degradation, release, and transport systems and several receptors that belong to the G protein-coupled receptor (GPCR) family. Targeting of this system with exogenous GHB is used in therapy to induce sleep and anesthesia and to reduce alcohol withdrawal syndrome. GHB is also popular as a recreational drug for its anxiolytic and mild euphoric effects. However, in both cases, GHB must be administered at high doses in order to maintain GHB concentrations in brain of ∼800-1,000 µM. These high concentrations are thought to be necessary for interactions with low-affinity sites on GABA(B) receptor, but the molecular targets and cellular mechanisms modulated by GHB remain poorly characterized. Therefore, to provide new insights into the elucidation of GHB mechanisms of action and open new tracks for future investigations, we explored changes of GHB-induced transcriptomes in rat hippocampus and prefrontal cortex by using DNA microarray studies. We demonstrate that a single acute anesthetic dose of 1 g/kg GHB alters a large number of genes, 121 in hippocampus and 53 in prefrontal cortex; 16 genes were modified simultaneously in both brain regions. In terms of molecular functions, the majority of modified genes coded for proteins or nucleotide binding sites. In terms of Gene Ontology (GO) functional categories, the largest groups were involved in metabolic processing for hippocampal genes and in biological regulation for prefrontal cortex genes. The majority of genes modified in both structures were implicated in cell communication processes. Western blot and immunohistochemical studies carried out on eight selected proteins confirmed the microarray findings.


Subject(s)
Central Nervous System Agents/pharmacology , Hippocampus/drug effects , Nerve Tissue Proteins/genetics , Prefrontal Cortex/drug effects , Sodium Oxybate/pharmacology , Animals , Blotting, Western , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Immunohistochemistry , Male , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Transcription, Genetic/drug effects
11.
Glia ; 58(2): 169-80, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19565659

ABSTRACT

Neurosteroids are synthesized either by glial cells, by neurons, or within the context of neuron-glia cross-talk. Various studies suggested neurosteroid involvement in the control of neurodegeneration but there is no evidence showing that the natural protection of nerve cells against apoptosis directly depends on their own capacity to produce neuroprotective neurosteroids. Here, we investigated the interactions between neurosteroidogenesis and apoptosis occurring in sensory structures of rats subjected to neuropathic pain generated by sciatic nerve chronic constriction injury (CCI). Using the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), we observed no apoptotic cells in the spinal cord up to 30 days after CCI although pain symptoms such as mechano-allodynia, thermal and mechanical hyperalgesia were evidenced with the Hargreaves's behavioral and von Frey filament tests. In contrast, double-labeling experiments combining TUNEL and immunostaining with antibodies against glutamine synthetase or neuronal nuclei protein revealed apoptosis occurrence in satellite glial cells (SGC) (not in neurons) of CCI rat ipsilateral dorsal root ganglia (DRG) at day 30 after injury. Pulse-chase experiments coupled with high performance liquid chromatography and flow scintillation detection showed that, among numerous biosynthetic pathways converting [(3)H]pregnenolone into various [(3)H]neurosteroids, only [(3)H]estradiol formation was selectively modified and upregulated in DRG of CCI rats. Consistently, immunohistochemical investigations localized aromatase (estradiol-synthesizing enzyme) in DRG neurons but not in SGC. Pharmacological inhibition of aromatase caused apoptosis of CCI rat DRG neurons. Altogether, our results suggest that endogenously produced neurosteroids such as estradiol may be pivotal for the protection of DRG sensory neurons against sciatic nerve CCI-induced apoptosis.


Subject(s)
Ganglia, Spinal/physiopathology , Neuroglia/physiology , Neurotransmitter Agents/metabolism , Pain/physiopathology , Sciatic Nerve/injuries , Sensory Receptor Cells/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , Cell Survival/drug effects , Estradiol/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/enzymology , Letrozole , Male , Neuroglia/drug effects , Neuroglia/enzymology , Nitriles/pharmacology , Pain/enzymology , Pain/etiology , Pain Measurement , Pregnenolone/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Nerve/physiopathology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/enzymology , Time Factors , Triazoles/pharmacology
12.
Psychoneuroendocrinology ; 34 Suppl 1: S169-77, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19577851

ABSTRACT

Neurotransmitters such as glutamate, substance P, serotonin and gamma-aminobutyric acid pivotally control pain mechanisms. It is also well known that inflammatory and/or neuropathic pain may depend on the action of diverse cytokines and other molecules including eicosanoids, endorphins, calcitonin-gene related peptide, free radicals and transcription factors. Because steroids control the development, activities and plasticity of the nervous system, these compounds are of particular interest in the modulation of pain. The paper discusses various data supporting the existence of key regulatory effects of steroids in the control of pain. In particular, we analyzed three categories of observations which historically contributed to demonstrate that endogenous and synthetic steroids play a crucial role in the regulation of neurobiological processes involved in pain sensation. The first series of data, which present the chemical characteristics enabling steroids to act on several tissues, also summarize pertinent results supporting the modulation of pain sensation by steroidal compounds. The second category of data evokes psychosocial, fundamental and clinical results suggesting the existence of sex steroid-based differences in pain perception. Finally, we discuss recent evidence showing the endogenous production of neurosteroids and their effects in the spinal cord which crucially controls pain transmission. Taken together, the data reviewed herein suggest that future investigations aiming to develop effective steroid-based strategies against chronic pain must integrate in a complementary manner anti-inflammatory properties of steroids, sex steroid-induced dimorphism in pain perception and regulatory effects exerted by endogenous neurosteroids in pain neural circuits.


Subject(s)
Gonadal Steroid Hormones/physiology , Neurotransmitter Agents/physiology , Pain/physiopathology , Steroids/physiology , Animals , Humans , Molecular Structure , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/therapeutic use , Pain/drug therapy , Sex Characteristics , Steroids/chemistry
13.
Neurobiol Dis ; 30(1): 30-41, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18291663

ABSTRACT

Identification of cellular targets pertinent for the development of effective therapies against pathological pain constitutes a difficult challenge. We combined several approaches to show that 3alpha-hydroxysteroid oxido-reductase (3alpha-HSOR), abundantly expressed in the spinal cord (SC), is a key target, the modulation of which markedly affects nociception. 3alpha-HSOR catalyzes the biosynthesis and oxidation of 3alpha,5alpha-reduced neurosteroids as allopregnanolone (3alpha,5alpha-THP), which stimulates GABA(A) receptors. Intrathecal injection of Provera (pharmacological inhibitor of 3alpha-HSOR activity) in naive rat SC decreased thermal and mechanical nociceptive thresholds assessed with behavioral methods. In contrast, pain thresholds were dose-dependently increased by 3alpha,5alpha-THP. In animals subjected to sciatic nerve injury-evoked neuropathic pain, molecular and biochemical experiments revealed an up-regulation of 3alpha-HSOR reductive activity in the SC. Enhancement of 3alpha,5alpha-THP concentration in the SC induced analgesia in neuropathic rats while Provera exacerbated their pathological state. Possibilities are opened for chronic pain control with drugs modulating 3alpha-HSOR activity in nerve cells.


Subject(s)
3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/metabolism , Pain Threshold/physiology , Sciatic Neuropathy/physiopathology , Spinal Cord/enzymology , Analysis of Variance , Animals , Behavior, Animal , Chromatography, High Pressure Liquid , Enzyme Inhibitors , Functional Laterality , Gene Expression Regulation, Enzymologic/drug effects , Hot Temperature , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , Medroxyprogesterone Acetate , Pain Measurement , Pain Threshold/drug effects , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Touch , Tritium/metabolism
14.
Neurochem Int ; 52(6): 948-55, 2008 May.
Article in English | MEDLINE | ID: mdl-18295935

ABSTRACT

Interactions between neurosteroidogenesis and proteins involved in age-related diseases are unknown. High concentrations of amyloid-beta (A beta) peptides induce plaques in Alzheimer's disease but several studies demonstrated that physiological or non-toxic doses are neuroprotective. We compared the effects of non-toxic and toxic concentrations of A beta 1-42 and A beta 25-35 on neurosteroidogenesis in human neuroblastoma SH-SY5Y cells. Viability assays revealed that nanomolar doses of A beta are devoid of cytotoxicity while 12 microM induced cell death. Pulse-chase, high-performance liquid chromatography and flow-scintillation analyses showed that non-toxic A beta 1-42 concentrations, acting selectively, decreased [3H]progesterone but increased [3H]estradiol production from the precursor [3H]pregnenolone. Non-toxic A beta 25-35 doses reduced [3H]progesterone formation but had no effect on [3H]estradiol biosynthesis. At 12 microM, both A beta 1-42 and A beta 25-35 inhibited [3H]progesterone formation but only A beta 1-42 reduced [3H]estradiol production. The results demonstrate a selective and amino-acid sequence-dependent action of A beta on neurosteroidogenesis. The fact that non-toxic A beta 1-42 doses stimulated neuroprotective-neurosteroid estradiol synthesis, which is inhibited by high A beta 1-42 doses, may explain A beta 1-42 ability to exert either protective or deleterious effects on nerve cells.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , Brain/metabolism , Neurons/metabolism , Steroids/biosynthesis , Alzheimer Disease/physiopathology , Amino Acid Sequence/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Brain/physiopathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Estrogens/biosynthesis , Humans , Neuroblastoma , Neurons/drug effects , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Pregnenolone/metabolism , Progesterone/biosynthesis
15.
FASEB J ; 22(1): 93-104, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17720801

ABSTRACT

We investigated the role and mechanism of action of dehydroepiandrosterone (DHEA) produced by the spinal cord (SC) in pain modulation in sciatic-neuropathic and control rats. Real-time polymerase chain reaction (PCR) after reverse transcription revealed cytochrome P450c17 (DHEA-synthesizing enzyme) gene repression in neuropathic rat SC. A combination of pulse-chase experiments, high performance liquid chromatography (HPLC), and flow-scintillation detection showed decreased DHEA biosynthesis from pregnenolone in neuropathic SC slices. Radioimmunoassays demonstrated endogenous DHEA level drop in neuropathic SC. Behavioral analysis showed a rapid pronociceptive and a delayed antinociceptive action of acute DHEA treatment. Inhibition of DHEA biosynthesis in the SC by intrathecally administered ketoconazole (P450c17 inhibitor) induced analgesia in neuropathic rats. BD1047 (sigma-1 receptor antagonist) blocked the transient pronociceptive effect evoked by acute DHEA administration. Chronic DHEA treatment increased and maintained elevated the basal nociceptive thresholds in neuropathic and control rats, suggesting that androgenic metabolites generated from daily administered DHEA exerted analgesic effects while DHEA itself (before being metabolized) induced a rapid pronociceptive action. Indeed, intrathecal administration of testosterone, an androgen deriving from DHEA, caused analgesia in neuropathic rats. Together, these molecular, biochemical, and functional results demonstrate that DHEA synthesized in the SC controls pain mechanisms. Possibilities are opened for pain modulation by drugs regulating P450c17 in nerve cells.


Subject(s)
Dehydroepiandrosterone/physiology , Pain/physiopathology , Spinal Cord/metabolism , Animals , Base Sequence , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , DNA Primers , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/pharmacology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Gene Expression , Injections, Spinal , Ketoconazole/administration & dosage , Ketoconazole/pharmacology , Male , Pain/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects
16.
J Neurobiol ; 66(8): 868-81, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16673391

ABSTRACT

Studies performed with animals suggest neurosteroid involvement in neuroprotection. However in humans, the role of neurosteroidogenesis in the regulation of degenerative processes is unknown. To determine whether cellular factors intervening in degenerative mechanisms may interfere with the process of neurosteroidogenesis in humans, we combined pulse-chase experiments with HPLC and continuous flow scintillation detection to compare neurosteroid production in normal and transfected SH-SY5Y cells with key proteins involved in Alzheimer's disease (AD). Microscope analyses revealed that cell morphology was unchanged in stably transfected SH-SY5Y cells overexpressing human native tau (hTau40), mutant tau (P301L), and wild-type amyloid precursor protein (APPwt) compared to controls. Biochemical investigations showed that hTau40 enhanced progesterone (PROG), 17OHPROG, testosterone, and 3alpha-androstanediol neosynthesis from pregnenolone. In contrast, tau with the pathogenic P301L mutation was devoid of action on neurosteroidogenesis. Overexpression of APPwt inhibited PROG formation, did not affect 17OHPROG and testosterone, but increased 3alpha-androstanediol and estradiol synthesis. Extracellular treatment of control cells with aggregated amyloid peptide mimicked the action of APPwt expression on PROG but not on 3alpha-androstanediol and estradiol production. Moreover, PROG biosynthesis in APPwt cells was up-regulated in the presence of a gamma-secretase inhibitor. Our results provide the first evidence for the regulation of neurosteroid biosynthesis by key proteins involved in the etiology of AD. The data suggest that pathogenic factors may induce neurodegeneration in humans through the reduction of the synthesis of endogenous neuroprotective neurosteroids in nerve cells.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Cytoprotection/physiology , Neurons/metabolism , Steroids/metabolism , tau Proteins/metabolism , 17-alpha-Hydroxyprogesterone/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Androstenediol/metabolism , Aspartic Acid Endopeptidases , Brain/metabolism , Brain/physiopathology , Cell Shape/genetics , Endopeptidases/drug effects , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Humans , Mutation/genetics , Neuroprotective Agents/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Progesterone/biosynthesis , Testosterone/biosynthesis , Transfection , Tumor Cells, Cultured , tau Proteins/genetics
18.
J Mol Neurosci ; 28(1): 17-31, 2006.
Article in English | MEDLINE | ID: mdl-16632873

ABSTRACT

The spinal cord (SC) is a biosynthetic center for neurosteroids, including pregnenolone (PREG), progesterone (PROG), and 3alpha/5alpha-tetrahydroprogesterone (3alpha/5alpha-THP). In particular, an active form of cytochrome P450 sidechain cleavage (P450scc) has been localized in sensory networks of the rat SC dorsal horn (DH). P450scc is the key enzyme catalyzing the conversion of cholesterol (CHOL) into PREG, the rate-limiting step in the biosynthesis of all classes of steroids. To determine whether neurosteroidogenesis might be involved in the pivotal role played by the DH in nociception, effects of neurogenic pain provoked by sciatic nerve ligature were investigated on P450scc expression, cellular distribution, and activity in the SC. P450scc mRNA concentration was threefold higher in the DH of neuropathic rats than in controls. The nerve ligature also increased the density of P450sccpositive neuronal perykarya and fibers in the ipsilateral DH. Incubation of spinal tissue homogenates with [3H]CHOL revealed that the amount of newly synthesized [3H]PREG from [3H]CHOLwas 80% higher in the DH of neuropathic rats. Radioimmunoassays showed an increase of PREG and 3alpha/5alpha-THP concentrations in neuropathic rat DH. The upregulation of PREG and 3alpha/5alpha-THP biosynthesis might be involved in endogenous mechanisms triggered by neuropathic rats to cope with the chronic pain state. 3alpha/5alpha-THP formation from PREG can also generate PROG, which decreases sensitivity to pain and protects nerve cells against degeneration. Because apoptotic cell death has been demonstrated in the DH during neuropathic pain, activation of neurosteroidogenesis in spinal tissues might also be correlated to the neuroprotective role of steroids in the SC.


Subject(s)
Pain/metabolism , Spinal Cord/metabolism , Steroids/biosynthesis , 3-Hydroxysteroid Dehydrogenases/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/metabolism , Animals , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Humans , Ligation , Neurons/cytology , Neurons/metabolism , Pregnenolone/metabolism , Progesterone/analogs & derivatives , Progesterone/metabolism , Sciatic Nerve/surgery , Spinal Cord/cytology , Steroid 17-alpha-Hydroxylase/metabolism
19.
Proc Natl Acad Sci U S A ; 102(25): 9044-9, 2005 Jun 21.
Article in English | MEDLINE | ID: mdl-15951421

ABSTRACT

A crucial biochemical reaction in vertebrates is progesterone conversion into neuroactive metabolites such as dihydroprogesterone (5alpha-DHP) and tetrahydroprogesterone (3alpha,5alpha-THP), which regulate several neurobiological processes, including stress, depression, neuroprotection, and analgesia. 3alpha,5alpha-THP is a potent stimulator of type A receptors of GABA, the main inhibitory neurotransmitter. Here, we show that in the spinal sensory circuit progesterone conversion into 5alpha-DHP and 3alpha,5alpha-THP is inhibited dose-dependently by substance P (SP), a major mediator of painful signals. We developed a triple-labeling approach coupled with multichannel confocal microscope analysis, which revealed that, in the spinal cord (SC), SP-releasing afferents project on sensory neurons expressing simultaneously neurokinin 1 receptors (rNK1) and key enzymes catalyzing progesterone metabolism. Evidence for a potent inhibitory effect of SP on 5alpha-DHP and 3alpha,5alpha-THP formation in the SC was provided by combining pulse-chase experiments using [3H]progesterone as precursor, HPLC, recrystallization of [3H]metabolites to constant specific activity, and continuous flow detection of radioactive steroids. The action of SP on progesterone metabolism was mimicked by the rNK1-specific agonist [Sar-9,Met(O2)11]-SP. The selective rNK1 antagonist SR140333 totally reversed the effect of SP on progesterone conversion into 5alpha-DHP and 3alpha,5alpha-THP. These results provide direct evidence for the occurrence of anatomical and functional interactions between the SP-rNK1 system and neuroactive steroid-producing cells in the SC. The data suggest that, through the local control of 3alpha,5alpha-THP concentration in spinal sensory circuit, the SP-rNK1 system may indirectly interfere with GABA(A) receptor activity in the modulation of nociceptive transmission.


Subject(s)
Neurons, Afferent/physiology , Nociceptors/physiology , Pain/physiopathology , Progesterone/metabolism , Spinal Cord/physiology , Substance P/pharmacology , Animals , Disease Models, Animal , Male , Models, Neurological , Neurons, Afferent/drug effects , Progesterone/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Substance P/analogs & derivatives , gamma-Aminobutyric Acid/physiology
20.
J Neurochem ; 93(5): 1220-30, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15934942

ABSTRACT

Various studies have indicated that exogenous dehydroepiandrosterone (DHEA) modulates several mechanisms in the CNS of rodents. As adult rodent glands do not secrete significant amounts of DHEA, its role as endogenous modulator of the CNS remains possible only if DHEA is produced by nerve cells. Therefore, the last decade has been marked by diverse unsuccessful investigations aiming to demonstrate the activity of cytochrome P450c17 (P450c17), the key DHEA-synthesizing enzyme, in adult rodent CNS. Here, we combined molecular, anatomical, cellular and neurochemical approaches to provide the first demonstration of the existence of P450c17 and bioactivity in adult rat spinal cord (SC). Real-time RT-PCR revealed P450c17 gene expression in all SC segments. Western blot analyses allowed identification of a specific P450c17 protein in the SC and immunohistochemical studies localized P450c17 in neurones and glial cells. Pulse-chase experiments combined with HPLC and radioactive steroid detection showed that SC slices converted [3H]pregnenolone into [3H]DHEA, a conversion markedly reduced by ketoconazole, a P450c17 inhibitor. Kinetics studies revealed accumulation of [3H]DHEA newly synthesized by SC slices in the incubation medium as its amount declined slowly. This first cellular mapping of an active P450c17 in adult rodent SC suggests that endogenous DHEA synthesized in spinal neural networks may control various spinally-mediated activities.


Subject(s)
Dehydroepiandrosterone/biosynthesis , Spinal Cord/metabolism , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Gene Expression , Immunohistochemistry , Male , Molecular Biology/methods , Neurochemistry/methods , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/cytology , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...