Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38481090

RESUMO

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.

2.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-34502138

RESUMO

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.


Assuntos
Arritmias Cardíacas/genética , Morte Súbita Cardíaca , Canal de Potássio ERG1/genética , Ativação do Canal Iônico , Potenciais de Ação , Arritmias Cardíacas/patologia , Células Cultivadas , Canal de Potássio ERG1/química , Canal de Potássio ERG1/metabolismo , Células HEK293 , Humanos , Masculino , Mutação de Sentido Incorreto , Domínios Proteicos , Transporte Proteico , Adulto Jovem
3.
Int J Mol Sci ; 22(13)2021 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-34203531

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Ácido Cinurênico/metabolismo , Xanturenatos/metabolismo , Animais , Dopamina/metabolismo , Cinurenina/metabolismo , Masculino , Ratos , Ratos Wistar
4.
Cells ; 10(3)2021 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-33801063

RESUMO

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.


Assuntos
Forma Celular , Microglia/citologia , Microglia/metabolismo , Neuroproteção , Neuroesteroides/metabolismo , Fagocitose , Pregnanolona/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Modelos Biológicos , Neuroproteção/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Fagocitose/efeitos dos fármacos , Pregnanolona/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Receptores de GABA/genética , Receptores de GABA/metabolismo , Soro
5.
Neurotox Res ; 38(1): 145-162, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32108299

RESUMO

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.


Assuntos
Analgésicos/administração & dosagem , Antineoplásicos/efeitos adversos , Modelos Animais de Doenças , Cloridrato de Duloxetina/administração & dosagem , Neuralgia/induzido quimicamente , Neuralgia/prevenção & controle , Pregnanolona/administração & dosagem , Animais , Docetaxel/efeitos adversos , Epirubicina/efeitos adversos , Feminino , Terapia Neoadjuvante/efeitos adversos , Neuralgia/fisiopatologia , Dor/induzido quimicamente , Dor/prevenção & controle , Medição da Dor , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/prevenção & controle , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/fisiopatologia
6.
Front Neuroendocrinol ; 55: 100795, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31562849

RESUMO

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.


Assuntos
Dor Crônica/metabolismo , Neuralgia/metabolismo , Neuroproteção/fisiologia , Neuroesteroides/metabolismo , Nociceptividade/fisiologia , Manejo da Dor , Dor Crônica/tratamento farmacológico , Humanos , Neuralgia/tratamento farmacológico , Neuroesteroides/farmacologia , Nociceptividade/efeitos dos fármacos
7.
J Neuroinflammation ; 16(1): 54, 2019 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-30825874

RESUMO

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.


Assuntos
Cloridrato de Fingolimode/farmacologia , Imunossupressores/farmacologia , Neurite Autoimune Experimental/patologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/patologia , Animais , Masculino , Neuritos/efeitos dos fármacos , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica , Ratos , Ratos Endogâmicos Lew , Receptores de Lisoesfingolipídeo/agonistas , Índice de Gravidade de Doença
8.
Cell Mol Neurobiol ; 39(4): 523-537, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30187261

RESUMO

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.


Assuntos
Neuralgia/metabolismo , Pregnanolona/metabolismo , Progesterona/metabolismo , Pesquisa Translacional Biomédica , Animais , Humanos , Modelos Biológicos , Neuralgia/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Pregnanolona/biossíntese , Progesterona/biossíntese , Progesterona/química
9.
Neurochem Int ; 118: 185-194, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29936187

RESUMO

Proteolipid protein (PLP) mutation causes oligodendrocyte degeneration and myelin disorders including Pelizaeus-Merzbacher Disease (PMD). As the pathophysiological mechanisms involved in PMD are poorly known, the development of therapies remains difficult. To elucidate the pathogenic pathways, an immortalized oligodendroglial cell line (158JP) expressing PLP mutation has been generated. Previous investigations revealed that 158JP oligodendrocytes exhibit several abnormalities including aberrant PLP insertion into the plasma membrane, cAMP, plasmalogen and cell cycle deficits. However, further clarifications of abnormal PLP-induced oligodendrocyte degeneration are required in order to identify relevant mechanisms to target for efficient protection against oligodendrocyte death. Because PLP overexpression may lead to its accumulation inside the endoplasmic reticulum (ER) and cause ER-stress, we explored whether ER-stress may pivotally determine 158JP cell survival/death. Viability assays, RT-qPCR, western blot and flow cytometry were combined to compare cell survival, ER-stress and apoptotic markers in 158JP and control (158N) oligodendrocytes. We observed a significant decreased viability/survival of 158JP compared to 158N cells. Consistently, ER-stress markers (BiP, caspase-12) increased in 158JP (+30%) compared to the controls. mRNA and protein ratios of apoptotic modulators (Bax/Bcl2) are higher in 158JP oligodendrocytes which are also more vulnerable than 158N cells to tunicamycin-induced ER-stress. Interestingly, 4-Phenylbutyrate (ER-stress inhibitor), which decreased ER-stress and apoptotic markers in 158JP cells, significantly increased their survival. Our results, which show a direct link between the viability and endogenous levels of ER-stress and apoptotic markers in 158JP cells, also suggest that 4-Phenylbutyrate-based strategy may contribute to develop effective strategies against oligodendrocyte dysfunctions/death and myelin disorders.


Assuntos
Citoproteção/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Mutação/fisiologia , Proteína Proteolipídica de Mielina/metabolismo , Oligodendroglia/metabolismo , Fenilbutiratos/farmacologia , Animais , Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Transformada , Citoproteção/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia
10.
J Neuroimmunol ; 278: 1-10, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25595246

RESUMO

Our objective was to develop a chronic model of EAN which could be used as a tool to test treatment strategies for CIDP. Lewis rats injected with S-palmitoylated P0(180-199) peptide developed a chronic, sometimes relapsing-remitting type of disease. Our model fulfills electrophysiological criteria of demyelination with axonal degeneration, confirmed by immunohistopathology. The late phase of the chronic disease was characterized by accumulation of IL-17(+) cells and macrophages in sciatic nerves and by high serum IL-17 levels. In conclusion, we have developed a reliable and reproducible animal model resembling CIDP that can now be used for translational drug studies.


Assuntos
Modelos Animais de Doenças , Proteína P0 da Mielina/química , Proteína P0 da Mielina/toxicidade , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/patologia , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/fisiopatologia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Eletrofisiologia , Potenciais Evocados/fisiologia , Interleucina-17/sangue , Linfócitos/metabolismo , Linfócitos/patologia , Macrófagos/patologia , Masculino , Peptídeos/toxicidade , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/sangue , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/induzido quimicamente , Ratos , Ratos Endogâmicos Lew , Nervo Isquiático/patologia , Nervo Isquiático/fisiopatologia , Linfócitos T/patologia
11.
PLoS One ; 8(11): e80915, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260511

RESUMO

Painful peripheral neuropathy belongs to major side-effects limiting cancer chemotherapy. Paclitaxel, widely used to treat several cancers, induces neurological symptoms including burning pain, allodynia, hyperalgesia and numbness. Therefore, identification of drugs that may effectively counteract paclitaxel-induced neuropathic symptoms is crucial. Here, we combined histopathological, neurochemical, behavioral and electrophysiological methods to investigate the natural neurosteroid 3α-androstanediol (3α-DIOL) ability to counteract paclitaxel-evoked peripheral nerve tissue damages and neurological symptoms. Prophylactic or corrective 3α-DIOL treatment (4 mg/kg/2 days) prevented or suppressed PAC-evoked heat-thermal hyperalgesia, cold-allodynia and mechanical allodynia/hyperalgesia, by reversing to normal, decreased thermal and mechanical pain thresholds of PAC-treated rats. Electrophysiological studies demonstrated that 3α-DIOL restored control values of nerve conduction velocity and action potential peak amplitude significantly altered by PAC-treatment. 3α-DIOL also repaired PAC-induced nerve damages by restoring normal neurofilament-200 level in peripheral axons and control amount of 2',3'-cyclic-nucleotide-3'-phosphodiesterase in myelin sheaths. Decreased density of intraepidermal nerve fibers evoked by PAC-therapy was also counteracted by 3α-DIOL treatment. More importantly, 3α-DIOL beneficial effects were not sedation-dependent but resulted from its neuroprotective ability, nerve tissue repairing capacity and long-term analgesic action. Altogether, our results showing that 3α-DIOL efficiently counteracted PAC-evoked painful symptoms, also offer interesting possibilities to develop neurosteroid-based strategies against chemotherapy-induced peripheral neuropathy. This article shows that the prophylactic or corrective treatment with 3α-androstanediol prevents or suppresses PAC-evoked painful symptoms and peripheral nerve dysfunctions in rats. The data suggest that 3α-androstanediol-based therapy may constitute an efficient strategy to explore in humans for the eradication of chemotherapy-induced peripheral neuropathy.


Assuntos
Androstano-3,17-diol/farmacologia , Antineoplásicos Fitogênicos/efeitos adversos , Hiperalgesia/prevenção & controle , Neuralgia/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Paclitaxel/efeitos adversos , Dor/prevenção & controle , Doenças do Sistema Nervoso Periférico/prevenção & controle , Potenciais de Ação/efeitos dos fármacos , Androstano-3,17-diol/análogos & derivados , Animais , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Masculino , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/patologia , Condução Nervosa/efeitos dos fármacos , Neuralgia/induzido quimicamente , Neuralgia/fisiopatologia , Dor/induzido quimicamente , Dor/fisiopatologia , Medição da Dor , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/patologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/fisiopatologia , Ratos , Ratos Sprague-Dawley
12.
Neuropharmacology ; 70: 254-60, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23422298

RESUMO

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.


Assuntos
Ácido Cinurênico/farmacologia , Neprilisina/biossíntese , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Indução Enzimática , Expressão Gênica/efeitos dos fármacos , Humanos , Ácido Cinurênico/análogos & derivados , Ácido Cinurênico/antagonistas & inibidores , Camundongos , Neprilisina/antagonistas & inibidores , Neprilisina/metabolismo , Fármacos Neuroprotetores/metabolismo , Cultura Primária de Células , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Tiorfano/farmacologia
13.
PLoS One ; 7(11): e48553, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23139790

RESUMO

Xanthurenic acid (XA) is a metabolite of the tryptophan oxidation pathway through kynurenine and 3-hydroxykynurenine. XA was until now considered as a detoxification compound and dead-end product reducing accumulation of reactive radical species. Apart from a specific role for XA in the signaling cascade resulting in gamete maturation in mosquitoes, nothing was known about its functions in other species including mammals. Based upon XA distribution, transport, accumulation and release in the rat brain, we have recently suggested that XA may potentially be involved in neurotransmission/neuromodulation, assuming that neurons presumably express specific XA receptors. Recently, it has been shown that XA could act as a positive allosteric ligand for class II metabotropic glutamate receptors. This finding reinforces the proposed signaling role of XA in brain. Our present results provide several lines of evidence in favor of the existence of specific receptors for XA in the brain. First, binding experiments combined with autoradiography and time-course analysis led to the characterization of XA binding sites in the rat brain. Second, specific kinetic and pharmacological properties exhibited by these binding sites are in favor of G-protein-coupled receptors (GPCR). Finally, in patch-clamp and calcium imaging experiments using NCB-20 cells that do not express glutamate-induced calcium signals, XA elicited specific responses involving activation of cationic channels and increases in intracellular Ca(2+) concentration. Altogether, these results suggest that XA, acting through a GPCR-induced cationic channel modulatory mechanism, may exert excitatory functions in various brain neuronal pathways.


Assuntos
Canais Iônicos/metabolismo , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Xanturenatos/metabolismo , Compostos de Anilina/metabolismo , Animais , Autorradiografia , Sítios de Ligação , Cálcio/metabolismo , Cátions , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Cinética , Masculino , Membranas/efeitos dos fármacos , Membranas/metabolismo , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurotransmissores/metabolismo , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Wistar , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Xantenos/metabolismo , Xanturenatos/química , Xanturenatos/farmacologia
14.
Pain ; 152(1): 170-181, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21071147

RESUMO

Oxaliplatin (OXAL) is a platinum-based drug used for the treatment of colorectal, lung, breast and ovarian cancers. OXAL does not cause renal or hematologic toxicity. However, OXAL induces neuropathic pain which hampers the chemotherapy success. Attempts with neuroprotective agents including anticonvulsivants and antidepressants were made to prevent OXAL-induced painful neuropathy but the clinical data are controversial and the tested neuroprotectors are able to evoke themselves undesirable effects. Here, we demonstrated that the natural neurosteroid allopregnanolone (3α,5α-THP), known to be devoid of toxic side-effects in humans and experimental models, prevented and suppressed OXAL-induced painful neuropathic symptoms. Indeed, 3α,5α-THP repaired OXAL-evoked neurochemical and functional alterations in peripheral nerves and intra-epidermal nerve fibers (IENF). Behavioral analyses showed that prophylactic or corrective 3α,5α-THP treatment (4mg/kg/2days) respectively prevented or abolished OXAL-induced cold allodynia, mechanical allodynia and hyperalgesia by reversing to normal decreased thermal and mechanical pain thresholds of OXAL-treated rats. Electrophysiological investigations revealed that 3α,5α-THP restored control values of sciatic nerve conduction velocity and action potential peak amplitude drastically reduced by OXAL-treatment. Furthermore, immunohistochemistry and confocal microscopic quantifications demonstrated that 3α,5α-THP repaired OXAL-induced neurochemical/cellular alterations by restoring IENF control density and normal level of neurofilament 200kDa that was strongly repressed by OXAL in dorsal root ganglion neurons and sciatic nerve axons. OXAL showed no toxicity for the non-compact myelin protein 2',3'-cyclic-nucleotide-3'-phosphodiesterase whose expression level was similarly increased by 3α,5α-THP in controls and OXAL-treated rat nerves. Together, these results may be interesting for the development of natural or safe neurosteroid-based neuroprotective strategy against anticancer drug-evoked painful neuropathy.


Assuntos
Anestésicos/uso terapêutico , Neuralgia/induzido quimicamente , Neuralgia/prevenção & controle , Compostos Organoplatínicos/efeitos adversos , Pregnanolona/uso terapêutico , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Animais , Modelos Animais de Doenças , Estimulação Elétrica , Comportamento Exploratório/efeitos dos fármacos , Gânglios Espinais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Masculino , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/fisiologia , Condução Nervosa/efeitos dos fármacos , Neuralgia/patologia , Proteínas de Neurofilamentos/metabolismo , Oxaliplatina , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Nervo Isquiático/fisiopatologia , Estatísticas não Paramétricas , Fatores de Tempo
15.
Cell Mol Life Sci ; 67(17): 3017-34, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20431905

RESUMO

Painful neuropathy is a major side-effect limiting cancer chemotherapy. Therefore, novel strategies are required to suppress the neuropathic effects of anticancer drugs without altering their chemotherapeutic effectiveness. By combining biochemical, neuroanatomical/neurochemical, electrophysiological and behavioral methods, we demonstrated that progesterone-derived neurosteroids including 5alpha-dihydroprogesterone and 3alpha,5alpha-tetrahydroprogesterone suppressed neuropathic symptoms evoked in naive rats by vincristine. Neurosteroids counteracted vincristine-induced alterations in peripheral nerves including 2',3'-cyclic nucleotide 3'-phosphodiesterase, neurofilament-200 kDa and intraepidermal nerve fiber repression, nerve conduction velocity, and pain transmission abnormalities (allodynia/hyperalgesia). In skin-tumor rats generated with carcinosarcoma-cells, vincristine, which suppressed the skin tumor and restored normal blood concentration of vascular endothelial growth factor (VEGF), reproduced neuropathic side-effects. Administered alone, neurosteroids did not affect the tumor and VEGF level. Combined with vincristine, neurosteroids preserved vincristine anti-tumor action but counteracted vincristine-induced neural side-effects. Together, these results provide valuable insight into the cellular and functional mechanisms underlying anticancer drug-induced neuropathy and suggest a neurosteroid-based strategy to eradicate painful neuropathy.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Neurotransmissores/uso terapêutico , Nervos Periféricos/efeitos dos fármacos , Vincristina/efeitos adversos , 5-alfa-Di-Hidroprogesterona/farmacologia , 5-alfa-Di-Hidroprogesterona/uso terapêutico , Análise de Variância , Animais , Linhagem Celular Tumoral , Eletrofisiologia , Hidroxiprogesteronas/farmacologia , Hidroxiprogesteronas/uso terapêutico , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Masculino , Condução Nervosa/fisiologia , Neurotransmissores/farmacologia , Medição da Dor , Nervos Periféricos/fisiologia , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas , Fator A de Crescimento do Endotélio Vascular/sangue
16.
Physiol Genomics ; 41(2): 146-60, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20103696

RESUMO

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.


Assuntos
Fármacos do Sistema Nervoso Central/farmacologia , Hipocampo/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Córtex Pré-Frontal/efeitos dos fármacos , Oxibato de Sódio/farmacologia , Animais , Western Blotting , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Proteínas do Tecido Nervoso/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Córtex Pré-Frontal/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Transcrição Gênica/efeitos dos fármacos
17.
Neuropharmacology ; 57(2): 137-47, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19427877

RESUMO

Several small chain fatty acids, including butyrate, valproate, phenylbutyrate and its derivatives, inhibit several HDAC activities in the brain at a several hundred micromolar concentration. Gamma-hydroxy-butyrate (GHB), a natural compound found in the brain originating from the metabolism of GABA, is structurally related to these fatty acids. The average physiological tissue concentration of GHB in the brain is below 50 microM, but when GHB is administered or absorbed for therapeutic or recreative purposes, its concentration reaches several hundred micromolars. In the present scenario, we demonstrate that pharmacological concentrations of GHB significantly induce brain histone H3 acetylation with a heterogeneous distribution in the brain and reduce in vitro HDAC activity. The degree of HDAC inhibition was also different according to the region of the brain considered. Taking into account the multiple physiological and functional roles attributed to the modification of histone acetylation and its consequences at the level of gene expression, we propose that part of the therapeutic or toxic effects of high concentrations of GHB in the brain after therapeutic administration of the drug could be partly due to GHB-induced epigenetic factors. In addition, we hypothesize that GHB, being naturally synthesized in the cytosolic compartment of certain neurons, could penetrate into the nuclei and may reach sufficient levels that could significantly modulate histone acetylation and may participate in the epigenetic modification of gene expression.


Assuntos
Encéfalo/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Inibidores de Histona Desacetilases , Histonas/metabolismo , Oxibato de Sódio/farmacologia , Acetilação/efeitos dos fármacos , Análise de Variância , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Cromatografia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Histona Desacetilases/metabolismo , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo
18.
PLoS One ; 3(2): e1641, 2008 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-18327293

RESUMO

BACKGROUND: Morphine, the principal active agent in opium, is not restricted to plants, but is also present in different animal tissues and cell types, including the mammalian brain. In fact, its biosynthetic pathway has been elucidated in a human neural cell line. These data suggest a role for morphine in brain physiology (e.g., neurotransmission), but this hypothesis remains a matter of debate. Recently, using the adrenal neuroendocrine chromaffin cell model, we have shown the presence of morphine-6-glucuronide (M6G) in secretory granules and their secretion products, leading us to propose that these endogenous alkaloids might represent new neuroendocrine factors. Here, we investigate the potential function of endogenous alkaloids in the central nervous system. METHODOLOGY AND PRINCIPAL FINDINGS: Microscopy, molecular biology, electrophysiology, and proteomic tools were applied to human neuroblastoma SH-SY5Y cells (i) to characterize morphine and M6G, and (ii) to demonstrate the presence of the UDP-glucuronyltransferase 2B7 enzyme, which is responsible for the formation of M6G from morphine. We show that morphine is secreted in response to nicotine stimulation via a Ca(2+)-dependent mechanism involving specific storage and release mechanisms. We also show that morphine and M6G at concentrations as low as 10(-10) M are able to evoke specific naloxone-reversible membrane currents, indicating possible autocrine/paracrine regulation in SH-SY5Y cells. Microscopy and proteomic approaches were employed to detect and quantify endogenous morphine in the mouse brain. Morphine is present in the hippocampus, cortex, olfactory bulb, and cerebellum at concentration ranging from 1.45 to 7.5 pmol/g. In the cerebellum, morphine immunoreactivity is localized to GABA basket cells and their termini, which form close contacts on Purkinje cell bodies. CONCLUSIONS/SIGNIFICANCE: The presence of morphine in the brain and its localization in particular areas lead us to conclude that it has a specific function in neuromodulation and/or neurotransmission. Furthermore, its presence in cerebellar basket cell termini suggests that morphine has signaling functions in Purkinje cells that remain to be discovered.


Assuntos
Química Encefálica , Cerebelo/química , Glucuronosiltransferase/análise , Derivados da Morfina/análise , Morfina/metabolismo , Alcaloides , Animais , Linhagem Celular Tumoral , Eletrofisiologia , Glucuronosiltransferase/metabolismo , Humanos , Camundongos , Neuroblastoma , Células de Purkinje , Transmissão Sináptica
19.
FASEB J ; 21(3): 885-95, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17197387

RESUMO

Two parent clones of a gamma-hydroxybutyrate (GHB) receptor, C12K32 and GHBh1, were isolated from a human frontal cortex cDNA library. The two clones differ by a deleted cytosine in C12K32. CHO cells transfected with either C12K32 or GHBh1 responded positively to submicromolar GHB stimulation. However, unlike C12K32, GHBh1 desensitizes rapidly on application of low concentrations of GHB. GHB receptor properties were then studied on C12K32 expressed in CHO cells. C12K32 bound GHB with a Kd of 114 nM and has no affinity for GABA or glutamate. GHB and NCS-382 displaced [3H]GHB with an IC50 of 53 +/- 8 and 120 +/- 18 nM, respectively. In patch-clamp experiments, GHB induced a dose-dependent response with an EC50 of 130 nM. This response was antagonized by NCS-382, was not reproduced by GABA, and was sensitive to the addition of GTP-gamma-S in the recording pipette. CHO cells transfected with C12K32 exhibited GTPgamma-35S binding with an EC50 of 462 nM for GHB and an IC50 of 2.9 microM for NCS-382. The present data led to the conclusion that both C12K32 and GHBh1 are two closely related isoforms of a human GHB receptor, GHBh1, that is described in the databank as the GPCR 172A.


Assuntos
Encéfalo/metabolismo , Receptores de Superfície Celular/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Humanos , Dados de Sequência Molecular , Ratos , Receptores de Superfície Celular/fisiologia , Receptores de GABA-B , Homologia de Sequência de Aminoácidos , Oxibato de Sódio/metabolismo
20.
J Comp Neurol ; 498(4): 508-24, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16874802

RESUMO

Gamma-hydroxybutyrate (GHB) is a substance derived from the metabolism of GABA and is heterogeneously distributed in various regions of the brain. This compound possesses a neuromodulatory role on several types of synapses, particularly those using GABA as a neurotransmitter. At physiological concentrations, this effect of GHB is mediated via specific receptors that induce neuronal hyperpolarization and bind radioactive GHB with a specific distribution, ontogenesis, kinetics, and pharmacology. A membrane protein that possesses six to seven transmembrane domains and which binds and is activated by micromolar amounts of GHB was recently cloned from rat brain hippocampus. In order to study the regional and cellular distribution of this receptor in rat brain, we selected several specific peptides belonging to the extracellular domains of the receptor to be used as specific immunogens to raise polyclonal antibodies in the rabbit. Among the antisera obtained, one of them gave particularly good results in terms of specificity and reactivity at high dilution. Immunohistochemical analyses, both at the confocal and electron microscopic level, showed receptor protein distribution closely resembling the distribution of GHB high-affinity binding sites, except for cerebellum, where GHB receptor(s) of lower affinity exist(s). In all regions studied the GHB receptor-like protein labeling appears to be distributed specifically in neurons and not in glial cells. At the cellular level the antibody specifically labels dendrites, and no immunoreactivity was detected in presynaptic endings or in axons. Accordingly, electron microscopy reveals strong labeling of postsynaptic densities and of neuronal cytosol.


Assuntos
Encéfalo/metabolismo , Receptores de Superfície Celular/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Encéfalo/fisiologia , Encéfalo/ultraestrutura , Células CHO , Cricetinae , DNA Complementar/análise , Eletrofisiologia , Hidroxibutiratos , Imuno-Histoquímica/métodos , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Distribuição Tecidual , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA