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










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167174, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631406

ABSTRACT

Mature oligodendrocytes (OLs) arise from oligodendrocyte precursor cells that, in case of demyelination, are recruited at the lesion site to remyelinate the axons and therefore restore the transmission of nerve impulses. It has been widely documented that exogenously administered steroid molecules are potent inducers of myelination. However, little is known about how neurosteroids produced de novo by OLs can impact this process. Here, we employed a human OL precursor cell line to investigate the role of de novo neurosteroidogenesis in the regulation of OLs differentiation, paying particular attention to the 18 kDa Translocator Protein (TSPO) which controls the rate-limiting step of the neurosteroidogenic process. Our results showed that, over the time of OL maturation, the availability of cholesterol, which is the neurosteroidogenesis initial substrate, and key members of the neurosteroidogenic machinery, including TSPO, were upregulated. In addition, OLs differentiation was impaired following neurosteroidogenesis inhibition and TSPO silencing. On the contrary, TSPO pharmacological stimulation promoted neurosteroidogenic function and positively impacted differentiation. Collectively, our results suggest that de novo neurosteroidogenesis is actively involved in the autocrine and paracrine regulation of human OL differentiation. Moreover, since TSPO was able to promote OL differentiation through a positive modulation of the neurosteroid biosynthetic process, it could be exploited as a promising target to tackle demyelinating diseases.


Subject(s)
Cell Differentiation , Oligodendroglia , Receptors, GABA , Humans , Receptors, GABA/metabolism , Receptors, GABA/genetics , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Oligodendroglia/cytology , Cell Differentiation/drug effects , Neurosteroids/metabolism , Cholesterol/metabolism , Cholesterol/biosynthesis , Cell Line , Myelin Sheath/metabolism
2.
J Neurochem ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38481090

ABSTRACT

Xanthurenic acid (XA) raises a growing multidisciplinary interest based upon its oxidizing properties, its ability to complex certain metal ions, and its detoxifier capacity of 3-hydroxykynurenine (3-HK), its brain precursor. However, little is still known about the role and mechanisms of action of XA in the central nervous system (CNS). Therefore, many research groups have recently investigated XA and its central functions extensively. The present paper critically reviews and discusses all major data related to XA properties and neuronal activities to contribute to the improvement of the current knowledge on XA's central roles and mechanisms of action. In particular, our data showed the existence of a specific G-protein-coupled receptor (GPCR) for XA localized exclusively in brain neurons exhibiting Ca2+ -dependent dendritic release and specific electrophysiological responses. XA properties and central activities suggest a role for this compound in brain intercellular signaling. Indeed, XA stimulates cerebral dopamine (DA) release contrary to its structural analog, kynurenic acid (KYNA). Thus, KYNA/XA ratio could be fundamental in the regulation of brain glutamate and DA release. Cerebral XA may also represent an homeostatic signal between the periphery and several brain regions where XA accumulates easily after peripheral administration. Therefore, XA status in certain psychoses or neurodegenerative diseases seems to be reinforced by its brain-specific properties in balance with its formation and peripheral inputs.

3.
Front Neuroendocrinol ; 72: 101116, 2024 01.
Article in English | MEDLINE | ID: mdl-38182090

ABSTRACT

The brain and spinal cord (SC) are both targeted by various hormones, including steroid hormones. However, investigations of the modulatory role of hormones on neurobiological functions usually focus only on the brain. The SC received little attention although this structure pivotally controls motor and sensory functions. Here, we critically reviewed key data showing that the process of neurosteroid biosynthesis or neurosteroidogenesis occurring in the SC plays a pivotal role in the modulation of peripheral nerve injury-induced chronic pain (PNICP) or neuropathic pain. Indeed, several active steroidogenic enzymes expressed in the SC produce endogenous neurosteroids that interact with receptors of neurotransmitters controlling pain. The spinal neurosteroidogenesis is differentially regulated during PNICP condition and its blockade modifies painful sensations. The paper suggests that future investigations aiming to develop effective strategies against PNICP or neuropathic pain must integrate in a gender or sex dependent manner the regulatory effects exerted by spinal neurosteroidogenesis.


Subject(s)
Chronic Pain , Neuralgia , Neurosteroids , Peripheral Nerve Injuries , Humans , Chronic Pain/etiology , Peripheral Nerve Injuries/complications , Spinal Cord , Neuralgia/etiology , Hormones
4.
Acta Neuropathol Commun ; 11(1): 56, 2023 03 31.
Article in English | MEDLINE | ID: mdl-37004127

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder with neuronal and synaptic losses due to the accumulation of toxic amyloid ß (Αß) peptide oligomers, plaques, and tangles containing tau (tubulin-associated unit) protein. While familial AD is caused by specific mutations, the sporadic disease is more common and appears to result from a complex chronic brain neuroinflammation with mitochondriopathies, inducing free radicals' accumulation. In aged brain, mutations in DNA and several unfolded proteins participate in a chronic amyloidosis response with a toxic effect on myelin sheath and axons, leading to cognitive deficits and dementia. Αß peptides are the most frequent form of toxic amyloid oligomers. Accumulations of misfolded proteins during several years alters different metabolic mechanisms, induce chronic inflammatory and immune responses with toxic consequences on neuronal cells. Myelin composition and architecture may appear to be an early target for the toxic activity of Aß peptides and others hydrophobic misfolded proteins. In this work, we describe the possible role of early myelin alterations in the genesis of neuronal alterations and the onset of symptomatology. We propose that some pathophysiological and clinical forms of the disease may arise from structural and metabolic disorders in the processes of myelination/demyelination of brain regions where the accumulation of non-functional toxic proteins is important. In these forms, the primacy of the deleterious role of amyloid peptides would be a matter of questioning and the initiating role of neuropathology would be primarily the fact of dysmyelination.


Subject(s)
Alzheimer Disease , Humans , Aged , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Myelin Sheath/metabolism , Axons/pathology , Neurons/metabolism
5.
Neurol Ther ; 11(3): 981-1042, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35610531

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) resulting in demyelination and neurodegeneration. The therapeutic strategy is now largely based on reducing inflammation with immunosuppressive drugs. Unfortunately, when disease progression is observed, no drug offers neuroprotection apart from its anti-inflammatory effect. In this review, we explore current knowledge on the assessment of neurodegeneration in MS and look at putative targets that might prove useful in protecting the axon from degeneration. Among them, Bruton's tyrosine kinase inhibitors, anti-apoptotic and antioxidant agents, sex hormones, statins, channel blockers, growth factors, and molecules preventing glutamate excitotoxicity have already been studied. Some of them have reached phase III clinical trials and carry a great message of hope for our patients with MS.

6.
Int J Mol Sci ; 23(5)2022 Mar 03.
Article in English | MEDLINE | ID: mdl-35269934

ABSTRACT

Neuromyelitis optica spectrum disorder (NMOSD) and multiple sclerosis (MS) are both autoimmune inflammatory and demyelinating diseases of the central nervous system. NMOSD is a highly disabling disease and rapid introduction of the appropriate treatment at the acute phase is crucial to prevent sequelae. Specific criteria were established in 2015 and provide keys to distinguish NMOSD and MS. One of the most reliable criteria for NMOSD diagnosis is detection in patient's serum of an antibody that attacks the water channel aquaporin-4 (AQP-4). Another target in NMOSD is myelin oligodendrocyte glycoprotein (MOG), delineating a new spectrum of diseases called MOG-associated diseases. Lastly, patients with NMOSD can be negative for both AQP-4 and MOG antibodies. At disease onset, NMOSD symptoms are very similar to MS symptoms from a clinical and radiological perspective. Thus, at first episode, given the urgency of starting the anti-inflammatory treatment, there is an unmet need to differentiate NMOSD subtypes from MS. Here, we used Fourier transform infrared spectroscopy in combination with a machine learning algorithm with the aim of distinguishing the infrared signatures of sera of a first episode of NMOSD from those of a first episode of relapsing-remitting MS, as well as from those of healthy subjects and patients with chronic inflammatory demyelinating polyneuropathy. Our results showed that NMOSD patients were distinguished from MS patients and healthy subjects with a sensitivity of 100% and a specificity of 100%. We also discuss the distinction between the different NMOSD serostatuses. The coupling of infrared spectroscopy of sera to machine learning is a promising cost-effective, rapid and reliable differential diagnosis tool capable of helping to gain valuable time in patients' treatment.


Subject(s)
Multiple Sclerosis , Neuromyelitis Optica , Aquaporin 4 , Autoantibodies , Humans , Machine Learning , Multiple Sclerosis/diagnosis , Myelin-Oligodendrocyte Glycoprotein
7.
Mol Neurobiol ; 59(3): 1744-1765, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35018577

ABSTRACT

Multiple sclerosis (MS) is an autoimmune and demyelinating disease of the central nervous system (CNS) caused by CNS infiltration of peripheral immune cells, immune-mediated attack of the myelin sheath, neuroinflammation, and/or axonal/neuronal dysfunctions. Some drugs are available to cope with relapsing-remitting MS (RRMS) but there is no therapy for the primary progressive MS (PPMS). Because growing evidence supports a regulatory role of the translocator protein (TSPO) in neuroinflammatory, demyelinating, and neurodegenerative processes, we investigated the therapeutic potential of phenylindolyilglyoxylamydes (PIGAs) TSPO ligands in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) mice mimicking the human PPMS. MOG-EAE C57Bl/6-mice were treated by TSPO ligands PIGA839, PIGA1138, or the vehicle. Several methods were combined to evaluate PIGAs-TSPO ligand effects on MOG-EAE symptoms, CNS infiltration by immune cells, demyelination, and axonal damages. PIGA1138 (15 mg/kg) drastically reduced MOG-EAE mice clinical scores, ameliorated motor dysfunctions assessed with the Catwalk device, and counteracted MOG-EAE-induced demyelination by preserving Myelin basic protein (MBP) expression in the CNS. Furthermore, PIGA1138-treatment prevented EAE-evoked decreased neurofilament-200 expression in spinal and cerebellar axons. Moreover, PIGA1138 inhibited peripheral immune-CD45 + cell infiltration in the CNS, suggesting that it may control inflammatory mechanisms involved in PPMS. Concordantly, PIGA1138 enhanced anti-inflammatory interleukin-10 serum level in MOG-EAE mice. PIGA1138-treatment, which increased neurosteroid allopregnanolone production, ameliorated all pathological biomarkers, while PIGA839, unable to activate neurosteroidogenesis in vivo, exerted only moderate/partial effects in MOG-EAE mice. Altogether, our results suggest that PIGA1138-based treatment may represent an interesting possibility to be explored for the innovation of effective therapies against PPMS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis, Chronic Progressive , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Ligands , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein
8.
Int J Mol Sci ; 22(17)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34502138

ABSTRACT

Long QT syndrome is one of the most common hereditary channelopathies inducing fatal arrhythmias and sudden cardiac death. We identified in a sudden arrhythmic death syndrome case a C-term KCNH2 mutation (c.3457C > T; p.His1153Tyr) classified as variant of unknown significance and functional impact. Heterologous expression in HEK293 cells combined with western-blot, flow-cytometry, immunocytochemical and microscope analyses shows no modification of channel trafficking to the cell membrane. Electrophysiological studies reveal that the mutation causes a loss of HERG channel function through an alteration of channel biophysical properties that reduces the current density leading to LQT2. These results provide the first functional evidence for H1153Y-KCNH2 mutation-induced abnormal channel properties. They concur with previous biophysical and clinical presentations of a survived patient with another variant that is G1036D. Therefore, the present report importantly highlights the potential severity of variants that may have useful implications for treatment, surveillance, and follow-up of LQT2 patients.


Subject(s)
Arrhythmias, Cardiac/genetics , Death, Sudden, Cardiac , ERG1 Potassium Channel/genetics , Ion Channel Gating , Action Potentials , Arrhythmias, Cardiac/pathology , Cells, Cultured , ERG1 Potassium Channel/chemistry , ERG1 Potassium Channel/metabolism , HEK293 Cells , Humans , Male , Mutation, Missense , Protein Domains , Protein Transport , Young Adult
9.
ACS Chem Neurosci ; 12(15): 2940-2945, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34292705

ABSTRACT

Hemes have been suggested to play a central role in Alzheimer's disease since they show high peroxidase reactivity when bound to amyloid ß peptides, leading to the production of reactive oxygen species. Here we used Fourier transform infrared and Raman imaging on Alzheimer's diseased mice and human brain tissue. Our finding suggests the accumulation of hemes in the senile plaques of both murine and human samples. We compared the Raman signature of the plaques to the ones of various hemeoproteins and to the hemin-Aß-42 complex. The detected Raman signature of the plaques does not allow identifying the type of heme accumulating in the plaques.


Subject(s)
Alzheimer Disease , Plaque, Amyloid , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Humans , Mice , Mice, Transgenic
10.
Int J Mol Sci ; 22(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203531

ABSTRACT

Xanthurenic acid (XA) is a metabolite of the kynurenine pathway (KP) synthetized in the brain from dietary or microbial tryptophan that crosses the blood-brain barrier through carrier-mediated transport. XA and kynurenic acid (KYNA) are two structurally related compounds of KP occurring at micromolar concentrations in the CNS and suspected to modulate some pathophysiological mechanisms of neuropsychiatric and/or neurodegenerative diseases. Particularly, various data including XA cerebral distribution (from 1 µM in olfactory bulbs and cerebellum to 0.1-0.4 µM in A9 and A10), its release, and interactions with G protein-dependent XA-receptor, glutamate transporter and metabotropic receptors, strongly support a signaling and/or neuromodulatory role for XA. However, while the parent molecule KYNA is considered as potentially involved in neuropsychiatric disorders because of its inhibitory action on dopamine release in the striatum, the effect of XA on brain dopaminergic activity remains unknown. Here, we demonstrate that acute local/microdialysis-infusions of XA dose-dependently stimulate dopamine release in the rat prefrontal cortex (four-fold increase in the presence of 20 µM XA). This stimulatory effect is blocked by XA-receptor antagonist NCS-486. Interestingly, our results show that the peripheral/intraperitoneal administration of XA, which has been proven to enhance intra-cerebral XA concentrations (about 200% increase after 50 mg/kg XA i.p), also induces a dose-dependent increase of dopamine release in the cortex and striatum. Furthermore, our in vivo electrophysiological studies reveal that the repeated/daily administrations of XA reduce by 43% the number of spontaneously firing dopaminergic neurons in the ventral tegmental area. In the substantia nigra, XA treatment does not change the number of firing neurons. Altogether, our results suggest that XA may contribute together with KYNA to generate a KYNA/XA ratio that may crucially determine the brain normal dopaminergic activity. Imbalance of this ratio may result in dopaminergic dysfunctions related to several brain disorders, including psychotic diseases and drug dependence.


Subject(s)
Brain/metabolism , Kynurenic Acid/metabolism , Xanthurenates/metabolism , Animals , Dopamine/metabolism , Kynurenine/metabolism , Male , Rats , Rats, Wistar
11.
Cells ; 10(3)2021 03 21.
Article in English | MEDLINE | ID: mdl-33801063

ABSTRACT

Microglial cells are key players in neural pathogenesis and microglial function regulation appears to be pivotal in controlling neuroinflammatory/neurological diseases. Here, we investigated the effects and mechanism of action of neurosteroid allopregnanolone (ALLO) on murine microglial BV-2 cells and primary microglia in order to determine ALLO-induced immunomodulatory potential and to provide new insights for the development of both natural and safe neuroprotective strategies targeting microglia. Indeed, ALLO-treatment is increasingly suggested as beneficial in various models of neurological disorders but the underlying mechanisms have not been elucidated. Therefore, the microglial cells were cultured with various serum concentrations to mimic the blood-brain-barrier rupture and to induce their activation. Proliferation, viability, RT-qPCR, phagocytosis, and morphology analyzes, as well as migration with time-lapse imaging and quantitative morphodynamic methods, were combined to investigate ALLO actions on microglia. BV-2 cells express subunits of GABA-A receptor that mediates ALLO activity. ALLO (10µM) induced microglial cell process extension and decreased migratory capacity. Interestingly, ALLO modulated the phagocytic activity of BV-2 cells and primary microglia. Our results, which show a direct effect of ALLO on microglial morphology and phagocytic function, suggest that the natural neurosteroid-based approach may contribute to developing effective strategies against neurological disorders that are evoked by microglia-related abnormalities.


Subject(s)
Cell Shape , Microglia/cytology , Microglia/metabolism , Neuroprotection , Neurosteroids/metabolism , Phagocytosis , Pregnanolone/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Shape/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation/drug effects , Humans , Mice, Inbred C57BL , Microglia/drug effects , Models, Biological , Neuroprotection/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Phagocytosis/drug effects , Pregnanolone/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA/genetics , Receptors, GABA/metabolism , Serum
12.
Trials ; 21(1): 591, 2020 Jun 29.
Article in English | MEDLINE | ID: mdl-32600454

ABSTRACT

BACKGROUND: Central nervous system damage in multiple sclerosis (MS) is responsible for serious deficiencies. Current therapies are focused on the treatment of inflammation; however, there is an urgent need for innovative therapies promoting neuroregeneration, particularly myelin repair. It is demonstrated that testosterone can act through neural androgen receptors and several clinical observations stimulated an interest in the potential protective effects of testosterone treatment for MS. Here, we sought to demonstrate the effects of a testosterone supplementation in testosterone-deficient men with relapsing-remitting MS. METHODS/DESIGN: This report presents the rationale and methodology of TOTEM RRMS, a French, phase 2, multicenter, randomized, placebo-controlled, and double-blind trial, which aims to prevent the progression of MS in men with low testosterone levels by administration of testosterone undecanoate, who were kept under natalizumab (Tysabri®) to overcome the anti-inflammatory effect of testosterone. Forty patients will be randomized into two groups receiving either a testosterone treatment (Nebido®) or a matching placebo. The intervention period for each group will last 66 weeks (treatment will be injected at baseline, week 6, and then every 12 weeks). The main objective is to determine the neuroprotective and remyelinating effects of testosterone using tensor diffusion imaging techniques and thalamic atrophy analyses. As secondary objectives, impacts of the testosterone supplementation will be studied using other conventional and unconventional MRI parameters and with clinical outcomes. DISCUSSION: The action of testosterone is observed in different experimental autoimmune encephalomyelitis models and epidemiological studies in humans. However, despite several preclinical data and some small clinical trials in MS, clear evidence for a therapeutic effect of hormone therapy is still missing. Therefore, our goal is to demonstrate the effects of testosterone therapies in MS. As there is no effective treatment currently available on fatigue in MS, careful attention should also be paid to secondary endpoints: fatigue, cognitive functions, and other symptoms that may improve life quality. Assuming a positive outcome of the trial, this treatment could be considered as a new neuroprotective and remyelinating therapy in relapsing-remitting MS and could be applicable to other demyelinating diseases. TRIAL REGISTRATION: ClinicalTrials.gov NCT03910738. Registered on 10 April 2019.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting/drug therapy , Myelin Sheath/drug effects , Testosterone/therapeutic use , Disease Progression , Double-Blind Method , Humans , Magnetic Resonance Imaging , Male , Multicenter Studies as Topic , Neuroprotection , Randomized Controlled Trials as Topic , Treatment Outcome
13.
Prog Neurobiol ; 190: 101800, 2020 07.
Article in English | MEDLINE | ID: mdl-32360535

ABSTRACT

Among several processes, a decrease in amyloid-beta (Aß) peptide elimination is thought to be one of the major pathophysiological factors in Alzheimer's disease (AD). Neprilysin (NEP) is a key metalloproteinase controlling the degradation and clearance of Aß peptides in the brain. NEP is induced by several pharmacological substances, amyloid deposits and somatostatin, but the physiological regulation of its expression remains unclear. This situation hampers the exploitation of NEP regulatory factors/mechanisms to develop effective strategies against Aß peptide accumulation-induced brain toxicity. Based on recent data aimed at elucidating this major question, the present paper addresses and critically discusses the role of 5-hydroxyindole-acetic acid (5-HIAA) and kynurenic acid (KYNA) in the regulation of NEP activity/expression in the brain. Both 5-HIAA and KYNA are endogenous metabolites of tryptophan, an essential amino-acid obtained through diet and gut microbiome. By interacting with the aryl hydrocarbon receptor, various tryptophan metabolites modulate several metalloproteinases regulating brain Aß peptide levels under normal and pathological conditions such as AD. In particular, interesting data reviewed here show that 5-HIAA and KYNA stimulate NEP activity/expression to prevent Aß peptide-induced neurotoxicity. These data open promising perspectives for the development of tryptophan metabolite-based therapies against AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hydroxyindoleacetic Acid/metabolism , Kynurenic Acid/metabolism , Neprilysin/metabolism , Tryptophan/metabolism , Humans
14.
Neurotox Res ; 38(1): 145-162, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32108299

ABSTRACT

Neoadjuvant chemotherapy is beneficial against breast cancer, but its toxicity causes painful chemotherapy-induced neuropathy which decreases seriously patients' quality of life. Development of effective therapy is crucial because current treatments are unsatisfactory. While animal models have previously been produced to test therapeutics against chemotherapy-induced neuropathy, neuropathic pain evoked by the frequently used neoadjuvant-chemotherapy involving sequentially epirubicin and docetaxel has never been modeled. Duloxetine, a serotonin/noradrenalin-reuptake inhibitor, is recommended against chemotherapy-induced neuropathy, but duloxetine exhibits controversial and adverse effects requiring its discontinuation. Here, we firstly produced and characterized a rat model for epirubicin-docetaxel induced painful neuropathy by using behavioral methods including the von Frey filament and the acetone tests that were combined with electrophysiological assessment of peripheral nerve functions and immunohistological analyzes. Using this model, we investigated the possibility to improve duloxetine efficacy and safety by combining its low doses (2 mg/kg/2 days) with the potent neuroprotector allopregnanolone (4 mg/kg/2 days). This concomitant therapy was more effective than separate duloxetine or allopregnanolone treatment to prevent epirubicin-docetaxel induced cold allodynia, mechanical allodynia/hyperalgesia, peripheral nerve functional/electrophysiological, and histological alterations. Interestingly, duloxetine-allopregnanolone concomitant treatment (but not duloxetine) also prevented epirubicin-docetaxel induced Schwann cell dedifferentiation and related macrophage (CD11b/c-positive cells) infiltration in sciatic nerves. Altogether, our results suggest that duloxetine and allopregnanolone concomitant treatment may represent a promising therapeutic option to counteract efficiently painful neuropathy or epirubicin-docetaxel evoked peripheral nerve tissue damages and dysfunctions.


Subject(s)
Analgesics/administration & dosage , Antineoplastic Agents/adverse effects , Disease Models, Animal , Duloxetine Hydrochloride/administration & dosage , Neuralgia/chemically induced , Neuralgia/prevention & control , Pregnanolone/administration & dosage , Animals , Docetaxel/adverse effects , Epirubicin/adverse effects , Female , Neoadjuvant Therapy/adverse effects , Neuralgia/physiopathology , Pain/chemically induced , Pain/prevention & control , Pain Measurement , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control , Sciatic Nerve/drug effects , Sciatic Nerve/physiopathology
15.
Front Neuroendocrinol ; 55: 100795, 2019 10.
Article in English | MEDLINE | ID: mdl-31562849

ABSTRACT

Complex mechanisms involved in neuropathic pain that represents a major health concern make its management complicated. Because neurosteroids are bioactive steroids endogenously synthesized in the nervous system, including in pain pathways, they appear relevant to develop effective treatments against neuropathic pain. Neurosteroids act in paracrine or autocrine manner through genomic mechanisms and/or via membrane receptors of neurotransmitters that pivotally modulate pain sensation. Basic studies which uncovered a direct link between neuropathic pain symptoms and endogenous neurosteroid production/regulation, paved the way for the investigations of neurosteroid therapeutic potential against pathological pain. Concordantly, antinociceptive properties of synthetic neurosteroids were evidenced in humans and animals. Neurosteroids promote peripheral analgesia mediated by T-type calcium and gamma-aminobutyric acid type A channels, counteract chemotherapy-induced neuropathic pain and ameliorate neuropathic symptoms of injured spinal cord animals by stimulating anti-inflammatory, remyelinating and neuroprotective processes. Together, these data open interesting perspectives for neurosteroid-based strategies to manage/alleviate efficiently neuropathic pain.


Subject(s)
Chronic Pain/metabolism , Neuralgia/metabolism , Neuroprotection/physiology , Neurosteroids/metabolism , Nociception/physiology , Pain Management , Chronic Pain/drug therapy , Humans , Neuralgia/drug therapy , Neurosteroids/pharmacology , Nociception/drug effects
16.
Analyst ; 144(15): 4647-4652, 2019 Aug 07.
Article in English | MEDLINE | ID: mdl-31257384

ABSTRACT

The challenging diagnosis and differentiation between multiple sclerosis and amyotrophic lateral sclerosis relies on the clinical assessment of the symptoms along with magnetic resonance imaging and sampling cerebrospinal fluid for the search of biomarkers for either disease. Despite the progress made in imaging techniques and biomarker identification, misdiagnosis still occurs. Here we used 2.5 µL of serum samples to obtain the infrared spectroscopic signatures of sera of multiple sclerosis and amyotrophic lateral sclerosis patients and compared them to those of healthy controls. The spectra are then classified with the help of a two-fold Random Forest cross-validation algorithm. This approach shows that infrared spectroscopy is powerful in discriminating between the two diseases and healthy controls by offering high specificity for multiple sclerosis (100%) and amyotrophic lateral sclerosis (98%). In addition, data after six and twelve months of treatment of the multiple sclerosis patients with biotin are discussed.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnosis , Biomarkers/blood , Multiple Sclerosis/diagnosis , Adult , Aged , Aged, 80 and over , Algorithms , Amyotrophic Lateral Sclerosis/drug therapy , Biotin/therapeutic use , Decision Trees , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Multiple Sclerosis/drug therapy , Pilot Projects , Spectroscopy, Fourier Transform Infrared/methods
17.
J Alzheimers Dis ; 72(4): 1045-1058, 2019.
Article in English | MEDLINE | ID: mdl-31256132

ABSTRACT

Translocator protein 18 kDa (TSPO) is located in the mitochondrial outer membrane and plays an important role in steroidogenesis and cell survival. In the central nervous system (CNS), its expression is upregulated in neuropathologies such as Alzheimer's disease (AD). Previously, we demonstrated that two new TSPO ligands based on an imidazoquinazolinone termed 2a and 2b, stimulated pregnenolone synthesis and ATP production in vitro. In the present study, we compared their effects to those of TSPO ligands described in the literature (XBD173, SSR-180,575, and Ro5-4864) by profiling the mitochondrial bioenergetic phenotype before and after treatment and investigating the protective effects of these ligands after oxidative injury in a cellular model of AD overexpressing amyloid-ß (Aß). Of note, ATP levels increased with rising pregnenolone levels suggesting that the energetic performance of mitochondria is linked to an increased production of this neurosteroid via TSPO modulation. Our results further demonstrate that the TSPO ligands 2a and 2b exerted neuroprotective effects by improving mitochondrial respiration, reducing reactive oxygen species and thereby decreasing oxidative stress-induced cell death as well as lowering Aß levels. The compounds 2a and 2b show similar or even better functional effects than those obtained with the reference TSPO ligands XBD173 and SSR-180.575. These findings indicate that the new TSPO ligands modulate mitochondrial bioenergetic phenotype and protect against oxidative injury probably through the de novo synthesis of neurosteroids, suggesting that these compounds could be potential new therapeutic tools for the treatment of neurodegenerative disease.


Subject(s)
Energy Metabolism/drug effects , Mitochondria/drug effects , Pregnenolone/biosynthesis , Quinazolinones/pharmacology , Receptors, GABA/metabolism , Cell Death/drug effects , Cell Line, Tumor , HEK293 Cells , Humans , Ligands , Mitochondria/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
18.
J Neuroinflammation ; 16(1): 54, 2019 Mar 02.
Article in English | MEDLINE | ID: mdl-30825874

ABSTRACT

BACKGROUND: Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an autoimmune-mediated inflammatory disease of the peripheral nervous system characterized by a response directed against certain myelin proteins and for which therapies are limited. Previous studies have suggested a beneficial role of FTY720, a sphingosine 1-phosphate (S1P) receptor agonist, known to deplete lymphocytes from the peripheral blood by sequestering them into lymph nodes, in the treatment of experimental autoimmune neuritis (EAN). Therefore, we investigated whether FTY720 is also beneficial in chronic experimental autoimmune neuritis (c-EAN), a recently developed rat model mimicking human CIDP. METHODS: c-EAN was induced in Lewis rats by immunization with S-palm P0(180-199) peptide. Rats were treated with FTY720 (1 mg/kg) or vehicle intraperitoneally once daily from the onset of clinical signs for 18 days; clinical signs were assessed daily until 60 days post-immunization (dpi). Electrophysiological and histological features were examined at different time points. We also evaluated the serum levels of different pro- and anti-inflammatory cytokines by ELISA or flow cytometry at 18, 40, and 60 dpi. RESULTS: Our data demonstrate that FTY720 decreased the severity and abolished the chronicity of the disease in c-EAN rats. Therapeutic FTY720 treatment reversed electrophysiological and histological anomalies, suggesting that myelinated fibers were subsequently preserved, it inhibited macrophage and IL-17+ cell infiltration in PNS, and it significantly reduced circulating pro-inflammatory cytokines. CONCLUSIONS: FTY720 treatment has beneficial effects on c-EAN, a new animal model mimicking human CIDP. We have shown that FTY720 is an effective immunomodulatory agent, improving the disease course of c-EAN, preserving the myelinated fibers, attenuating the axonal degeneration, and decreasing the number of infiltrated inflammatory cells in peripheral nerves. These data confirm the interest of testing FTY720 or molecules targeting S1P in human peripheral neuropathies.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/pharmacology , Neuritis, Autoimmune, Experimental/pathology , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Animals , Male , Neurites/drug effects , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Rats , Rats, Inbred Lew , Receptors, Lysosphingolipid/agonists , Severity of Illness Index
19.
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
20.
Acta Neuropathol Commun ; 6(1): 136, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30537985

ABSTRACT

Serotoninergic activation which decreases brain Aß peptides is considered beneficial in mouse models for Alzheimer's disease (AD), but the mechanisms involved remain unclear. Because growing evidence suggested that the stimulation of proteases digesting Aß, especially the endopeptidase neprilysin (NEP) may be effective for AD therapy/prevention, we explored the involvement of serotonin precursors and derivatives in NEP regulation. We found that 5-hydroxyindolacetic acid (5-HIAA), the final metabolite of serotonin, considered until now as a dead-end and inactive product of serotonin catabolism, significantly reduces brain Aß in the transgenic APPSWE mouse model for AD-related Aß pathology and in the phosphoramidon-induced cerebral NEP inhibition mouse model. 5-HIAA treatment improves memory performance in APPSWE mice. Furthermore, 5-HIAA and its precursors increase NEP level in vivo and in neuroblastoma cells. Inhibition of ERK 1/2 cascade by 5-HIAA or SCH772984 enhanced NEP levels, suggesting MAP-kinase pathway involvement in 5-HIAA-induced regulation of NEP expression. Our results provide the first demonstration that 5-HIAA is an active serotonin metabolite that increases brain Aß degradation/clearance and improves symptoms in the APPSWE mouse model for AD.


Subject(s)
Alzheimer Disease , Antipsychotic Agents/therapeutic use , Brain/metabolism , Gene Expression Regulation/drug effects , Hydroxyindoleacetic Acid/therapeutic use , Neprilysin/metabolism , 5-Hydroxytryptophan/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Cell Line, Tumor , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/physiology , Glycopeptides/therapeutic use , Humans , Male , Mice , Mice, Transgenic , Neprilysin/genetics , Signal Transduction/drug effects , Spatial Behavior/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...