Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
Behav Brain Res ; 416: 113557, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34453973

RESUMO

Ethanol withdrawal commonly leads to anxiety-related disorder, a central factor toward negative reinforcement leading to relapse. The lateral habenula (LHb), an epithalamic nucleus, has emerged to be critical for both reward and aversion processing. Recent studies have also implicated the hyperactivity of LHb, adding to the emergence of negative emotional states during withdrawal from addictive drugs. Herein, we have studied the effects of glutamate transporter inhibitor (PDC), GluN2B-containing NMDAR antagonist (Ro25-6981), and intracellular calcium chelator (BAPTA-AM) injection in LHb on ethanol withdrawal symptoms. We found that ethanol 4 g/kg 20 % w/v intragastric (i.g.) for 10 days followed by 24 h of withdrawal showed a significant increase in somatic signs characterized by vocalization, shaking, and scratching. It also increased locomotor activity and anxiety-like behavior, collectively showing expression of ethanol withdrawal symptoms. The intra-LHb administration of PDC (0.5 ng) worsened the effect of ethanol withdrawal, whereas Ro25-6981 (2 and 4 ng) and BAPTA-AM (6.5 and 13 ng) significantly reversed ethanol withdrawal-induced behavior evident by a decrease in somatic signs, locomotor activity, and anxiety-like behavior. Further, pretreatment of Ro25-6981 and BAPTA-AM reduced the neuronal loss, whereas PDC increased it compared to the vehicle-treated group, as evidenced by NeuN staining. Altogether, our results suggest that increased glutamate, GluN2B activation, and likely calcium increase indicative of glutamate excitotoxicity-induced neuronal loss in LHb possibly endorse the emergence of ethanol withdrawal symptoms, while their inhibition might help in alleviating the ethanol withdrawal symptoms.


Assuntos
Etanol/farmacologia , Ácido Glutâmico/toxicidade , Habenula/efeitos dos fármacos , Fenóis/farmacologia , Piperidinas/farmacologia , Síndrome de Abstinência a Substâncias , Alcoolismo/metabolismo , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Ansiedade , Masculino , Camundongos , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/fisiopatologia
2.
J Am Chem Soc ; 143(3): 1513-1520, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33449695

RESUMO

Photopharmacology addresses the challenge of drug selectivity and side effects through creation of photoresponsive molecules activated with light with high spatiotemporal precision. This is achieved through incorporation of molecular photoswitches and photocages into the pharmacophore. However, the structural basis for the light-induced modulation of inhibitory potency in general is still missing, which poses a major design challenge for this emerging field of research. Here we solved crystal structures of the glutamate transporter homologue GltTk in complex with photoresponsive transport inhibitors-azobenzene derivative of TBOA (both in trans and cis configuration) and with the photocaged compound ONB-hydroxyaspartate. The essential role of glutamate transporters in the functioning of the central nervous system renders them potential therapeutic targets in the treatment of neurodegenerative diseases. The obtained structures provide a clear structural insight into the origins of photocontrol in photopharmacology and lay the foundation for application of photocontrolled ligands to study the transporter dynamics by using time-resolved X-ray crystallography.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Compostos Azo/metabolismo , Sistema X-AG de Transporte de Aminoácidos/química , Ácido Aspártico/efeitos da radiação , Compostos Azo/química , Compostos Azo/efeitos da radiação , Cristalografia por Raios X , Ligação Proteica , Estereoisomerismo , Thermococcus/química , Raios Ultravioleta
3.
Neuroimage ; 225: 117457, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33069862

RESUMO

Functional MRI responses are localized to the synaptic sites of evoked inhibitory neurons, but it is unknown whether, or by what mechanisms, these neurons initiate functional hyperemia. Here, the neuronal origins of these hemodynamic responses were investigated by fMRI or local field potential and blood flow measurements during topical application of pharmacological agents when GABAergic granule cells in the rat olfactory bulb were synaptically targeted. First, to examine if postsynaptic activation of these inhibitory neurons was required for neurovascular coupling, we applied an NMDA receptor antagonist during cerebral blood volume-weighted fMRI acquisition and found that responses below the drug application site (up to ~1.5 mm) significantly decreased within ~30 min. Similarly, large decreases in granule cell postsynaptic activities and blood flow responses were observed when AMPA or NMDA receptor antagonists were applied. Second, inhibition of nitric oxide synthase preferentially decreased the initial, fast component of the blood flow response, while inhibitors of astrocyte-specific glutamate transporters and vasoactive intestinal peptide receptors did not decrease blood flow responses. Third, inhibition of GABA release with a presynaptic GABAB receptor agonist caused less reduction of neuronal and blood flow responses compared to the postsynaptic glutamate receptor antagonists. In conclusion, local hyperemia by synaptically-evoked inhibitory neurons was primarily driven by their postsynaptic activities, possibly through NMDA receptor-dependent calcium signaling that was not wholly dependent on nitric oxide.


Assuntos
Encéfalo/diagnóstico por imagem , Circulação Cerebrovascular/fisiologia , Neurônios GABAérgicos/fisiologia , Acoplamento Neurovascular/fisiologia , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Encéfalo/fisiologia , Circulação Cerebrovascular/efeitos dos fármacos , Estimulação Elétrica , Neuroimagem Funcional , Agonistas dos Receptores de GABA-B , Neurônios GABAérgicos/efeitos dos fármacos , Fluxometria por Laser-Doppler , Imageamento por Ressonância Magnética , Inibição Neural , Acoplamento Neurovascular/efeitos dos fármacos , Óxido Nítrico Sintase/antagonistas & inibidores , Bulbo Olfatório/citologia , Ratos , Receptores de AMPA/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de Peptídeo Intestinal Vasoativo/antagonistas & inibidores
4.
J Biol Chem ; 295(13): 4359-4366, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32079674

RESUMO

Excitatory amino acid transporters (EAATs) represent a protein family that is an emerging drug target with great therapeutic potential for managing central nervous system disorders characterized by dysregulation of glutamatergic neurotransmission. As such, it is of significant interest to discover selective modulators of EAAT2 function. Here, we applied computational methods to identify specific EAAT2 inhibitors. Utilizing a homology model of human EAAT2, we identified a binding pocket at the interface of the transport and trimerization domain. We next conducted a high-throughput virtual screen against this site and identified a selective class of EAAT2 inhibitors that were tested in glutamate uptake and whole-cell electrophysiology assays. These compounds represent potentially useful pharmacological tools suitable for further exploration of the therapeutic potential of EAAT2 and may provide molecular insights into mechanisms of allosteric modulation for glutamate transporters.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Doenças do Sistema Nervoso Central/tratamento farmacológico , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/química , Sistema X-AG de Transporte de Aminoácidos/genética , Animais , Sítios de Ligação/genética , Transporte Biológico/efeitos dos fármacos , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/patologia , Biologia Computacional , Transportador 2 de Aminoácido Excitatório/química , Transportador 2 de Aminoácido Excitatório/genética , Humanos , Ligação Proteica/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Interface Usuário-Computador
5.
Cell Rep ; 27(10): 2871-2880.e2, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31167134

RESUMO

Retinal waves, the spontaneous patterned neural activities propagating among developing retinal ganglion cells (RGCs), instruct the activity-dependent refinement of visuotopic maps. Although it is known that the wave is initiated successively by amacrine cells and bipolar cells, the behavior and function of glia in retinal waves remain unclear. Using multiple in vivo methods in larval zebrafish, we found that Müller glial cells (MGCs) display wave-like spontaneous activities, which start at MGC processes within the inner plexiform layer, vertically spread to their somata and endfeet, and horizontally propagate into neighboring MGCs. MGC waves depend on glutamatergic signaling derived from bipolar cells. Moreover, MGCs express both glia-specific glutamate transporters and the AMPA subtype of glutamate receptors. The AMPA receptors mediate MGC calcium activities during retinal waves, whereas the glutamate transporters modulate the occurrence of retinal waves. Thus, MGCs can sense and regulate retinal waves via AMPA receptors and glutamate transporters, respectively.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Cálcio/metabolismo , Células Ependimogliais/metabolismo , Ácido Glutâmico/metabolismo , Neuroglia/metabolismo , Receptores de AMPA/metabolismo , Células Ganglionares da Retina/metabolismo , Células Amácrinas/metabolismo , Células Amácrinas/fisiologia , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/genética , Animais , Animais Geneticamente Modificados , Células Ependimogliais/citologia , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/fisiologia , Ácido Glutâmico/farmacologia , Larva/efeitos dos fármacos , Larva/metabolismo , Larva/fisiologia , Neuroglia/citologia , Neuroglia/fisiologia , Receptores de AMPA/antagonistas & inibidores , Retina/citologia , Retina/metabolismo , Retina/fisiologia , Células Bipolares da Retina/metabolismo , Células Bipolares da Retina/fisiologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/fisiologia , Peixe-Zebra
6.
ACS Chem Biol ; 14(5): 1002-1010, 2019 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-31026143

RESUMO

Plasma-membrane glutamate transporters of the excitatory amino acid transporter (EAAT) family are important for maintaining a low glutamate concentration in the extracellular space of the mammalian brain. Glutamate is believed to be transported in its negatively charged form and energetically driven by the cotransport of three sodium ions, at least two of which are bound within the dielectric of the membrane. It was hypothesized that binding of substrates and competitive inhibitors is also electrogenic because the binding site is located near the center of the membrane. To test this hypothesis, we rapidly applied a low-affinity competitive inhibitor, kainate, to the glutamate transporter subtype EAAT2, resulting in outward transient current caused by movement of net negative charge of the inhibitor into the low dielectric of the protein/membrane. Consistent with these data, rate constants for inhibitor dissociation and binding were also voltage dependent. Our results are supported by electrostatic calculations and molecular dynamics simulations of spontaneous substrate dissociation, showing that the substrate and inhibitor binding site is located within the membrane environment of low dielectric constant. Charge movement caused by binding of negatively charged amino acid substrate is compensated by the charge of cotransported Na+ ion(s), thus preventing inhibition of substrate binding at negative membrane potentials. This charge compensation mechanism may be relevant for other Na+-driven transporters which recognize negatively charged substrates.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Fenômenos Biofísicos , Encéfalo/metabolismo , Ácido Caínico/metabolismo , Cinética , Mamíferos , Potenciais da Membrana , Simulação de Dinâmica Molecular , Especificidade por Substrato
7.
Physiol Behav ; 199: 28-32, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30389478

RESUMO

Glutamate transporter GLT1 mediates glutamate uptake, and maintains glutamate homeostasis in the synaptic cleft. Previous studies suggest that blockade of glutamate uptake affects synaptic transmission and plasticity. However, the effect of GLT1 blockade on learning and memory still receives little attention. In the present study, we examined the effect of unilateral intracerebroventricular injection of dihydrokainic acid (DHK), a GLT-1 inhibitor, on novel object recognition (NOR) memory performance. The NOR task involved three sessions including habituation, sampling and test. In experiment 1, DHK injection 0.5 h pre-sampling impaired short-term NOR memory performance. In experiment 2, DHK injection 0.5 h pre-sampling impaired long-term NOR memory acquisition. In experiment 3, DHK injection immediately but not 6 h post-sampling impaired long-term NOR memory consolidation. In experiment 4, DHK injection 0.5 h pre-test impaired long-term NOR memory retrieval. Furthermore, DHK-induced memory performance impairment was not due to its effects on nonspecific responses such as locomotor activity and exploratory behavior. The current findings further extend previous studies on the effects of disruption of glutamate homeostasis on learning and memory.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Ácido Caínico/análogos & derivados , Consolidação da Memória/efeitos dos fármacos , Reconhecimento Psicológico/efeitos dos fármacos , Animais , Ácido Caínico/farmacologia , Masculino , Camundongos
8.
J Pharmacol Sci ; 138(3): 176-183, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30322803

RESUMO

We previously reported that systemic administration of a delta opioid receptor (DOP) agonist, KNT-127, produced a potent anxiolytic-like effect in rats. Interestingly, DOPs are highly distributed in the prelimbic medial prefrontal cortex (PL-PFC). In the present study, we investigated the effect of KNT-127 co-perfusion in the PL-PFC on anxiety-like behavior in mice, induced by a glial glutamate transporter inhibitor, (3S)-3-[[3-[[4-(Trifluoromethyl)benzoyl]amino]phenyl]methoxy]-l-aspartic acid (TFB-TBOA). Extracellular glutamate levels were measured in male C57BL/6N mice by in vivo microdialysis high-performance liquid chromatography/electrochemical detection, with behavior simultaneously assessed in the open field test. As expected, extracellular glutamate levels were significantly increased, and anxiety-like behavior was induced after local perfusion of TFB-TBOA in the PL-PFC. Uniquely, co-perfusion of KNT-127 in the PL-PFC diminished anxiety-like behavior induced by TFB-TBOA without affecting extracellular glutamate levels. Further, the effect of KNT-127 on anxiety-like behavior was antagonized by a selective DOP antagonist, naltrindole, suggesting that KNT-127 acts via DOPs. These findings do not support our preconceived hypothesis that KNT-127 in PL-PFC produces an anxiolytic-like effect via suppression of glutamatergic transmission. Hence, further studies are necessary to understand the mechanisms of DOP agonist-induced anxiolytic-like effects in the PL-PFC.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Ansiolíticos/farmacologia , Ansiedade/induzido quimicamente , Ansiedade/prevenção & controle , Ácido Aspártico/análogos & derivados , Morfinanos/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptores Opioides delta/agonistas , Animais , Ansiolíticos/administração & dosagem , Ácido Aspártico/administração & dosagem , Ácido Aspártico/antagonistas & inibidores , Ácido Aspártico/farmacologia , Interações Medicamentosas , Ácido Glutâmico/metabolismo , Masculino , Camundongos , Microinjeções , Morfinanos/administração & dosagem , Morfinanos/antagonistas & inibidores , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Ácido gama-Aminobutírico/metabolismo
9.
Ther Hypothermia Temp Manag ; 8(3): 143-149, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29420129

RESUMO

Deep and profound hypothermia is successfully practiced in the prevention of ischemic stroke consequences and aortic arch cardiac surgery accompanied by reduction of cerebral circulation. Hypothermia is a current neuroprotection standard in hypoxic/ischemic encephalopathy. Drug-hypothermia administration is proposed as a new approach in pharmacotherapy for neonatal seizures. Also, hypothermia is useful as neuroprotective approach in long-term interplanetary space missions. We recently revealed gradual dynamics of hypothermia-induced decrease in transporter-mediated release and uptake of L-[14C]glutamate in presynaptic rat brain nerve terminals (synaptosomes), thereby confirming potent unspecific neuroprotective effect of hypothermia. Glutamate homo- and heteroexchange are significant mechanisms involved in the maintenance of the extracellular glutamate level in nerve terminals. We have analyzed whether glutamate homo- and heteroexchange in nerve terminals is temperature sensitive. In this study we showed that synaptosomal glutamate-induced L-[14C]glutamate release (homoexchange) and D-aspartate- and DL-threo-ß-hydroxyaspartate-induced L-[14C]glutamate release (heteroexchange) gradually decreased from deep (27°C) to profound (17°C) hypothermia with dynamics similar to that of glutamate transporter reversal. Interestingly, ambient L-[14C]glutamate concentration in the nerve terminal preparations remained unaltered during hypothermia administration. Therefore, we demonstrated that glutamate homo- and heteroexchange decreased from deep to profound hypothermia thereby preventing further elevation of extracellular glutamate. Hypothermia uncovered the principal processes contributing to glutamate homo- and heteroexchange in nerve terminals and the maintenance of definite ambient glutamate concentration. Additionally, we showed that glutamate transporter reversal can be nonpathological and occurs under physiological conditions at least as a part of homo- and heteroexchange mechanisms.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Ácido Glutâmico/metabolismo , Hipotermia/metabolismo , Terminações Pré-Sinápticas/metabolismo , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Radioisótopos de Carbono , Hipotermia Induzida , Técnicas In Vitro , Masculino , Ratos , Ratos Wistar
10.
Brain Res ; 1678: 129-137, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29066369

RESUMO

In Alzheimer's disease (AD), dementia severity correlates most strongly with decreased synapse density in the hippocampus and cerebral cortex. Although studies in rodents have established that hippocampal long-term potentiation (LTP) is inhibited by soluble oligomers of beta-amyloid (Aß), the synaptic mechanisms remain unclear. Here, field excitatory postsynaptic potentials (fEPSP) recordings were made in the CA1 region of mouse hippocampal slices. The medium of APP-expressing CHO cells, which contain soluble forms of Aß including small oligomers, inhibited LTP and facilitated long-term depression (LTD), thus making the LTP/LTD curve shift toward the right. This phenomenon could be mimicked by the non-selective glutamate transporter inhibitor, DL-TBOA. More specifically, the Aß impaired LTP and facilitated LTD were occluded by the selective astrocytic glutamate transporter inhibitors, TFB-TBOA. In cultured astrocytes, the Aß oligomers also decrease astrocytic glutamate transporters (EAAT1, EAAT2) expression. We conclude that soluble Aß oligomers decrease the activation of astrocytic glutamate transporters, thereby impairing synaptic plasticity.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia , Doença de Alzheimer/metabolismo , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Animais , Astrócitos/metabolismo , Região CA1 Hipocampal/metabolismo , Células CHO , Cricetulus , Fármacos Atuantes sobre Aminoácidos Excitatórios/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/metabolismo
11.
Org Biomol Chem ; 15(11): 2341-2344, 2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-28244539

RESUMO

The complex amino acid (l-threo)-3-[3-[4-(trifluoromethyl)benzoylamino]benzyloxy]aspartate (l-TFB-TBOA) and its derivatives are privileged compounds for studying the roles of excitatory amino acid transporters (EAATs) in regulation of glutamatergic neurotransmission, animal behavior, and in the pathogenesis of neurological diseases. The wide-spread use of l-TFB-TBOA stems from its high potency of EAAT inhibition and the lack of off-target binding to glutamate receptors. However, one of the main challenges in the evaluation of l-TFB-TBOA and its derivatives is the laborious synthesis of these compounds in stereoisomerically pure form. Here, we report an efficient and step-economic chemoenzymatic route that gives access to enantio- and diastereopure l-TFB-TBOA and its derivatives at multigram scale.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Aminoácidos/síntese química , Aminoácidos/metabolismo , Ácido Aspártico/análogos & derivados , Enzimas/metabolismo , Aminoácidos/química , Ácido Aspártico/síntese química , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Estrutura Molecular , Estereoisomerismo
12.
Glia ; 65(2): 293-308, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27785828

RESUMO

Perivascular endfeet of astrocytes are highly polarized compartments that ensheath blood vessels and contribute to the blood-brain barrier. They experience calcium transients with neuronal activity, a phenomenon involved in neurovascular coupling. Endfeet also mediate the uptake of glucose from the blood, a process stimulated in active brain regions. Here, we demonstrate in mouse hippocampal tissue slices that endfeet undergo sodium signaling upon stimulation of glutamatergic synaptic activity. Glutamate-induced endfeet sodium transients were diminished by TFB-TBOA, suggesting that they were generated by sodium-dependent glutamate uptake. With local agonist application, they could be restricted to endfeet and immunohistochemical analysis revealed prominent expression of glutamate transporters GLAST and GLT-1 localized towards the neuropil vs. the vascular side of endfeet. Endfeet sodium signals spread at an apparent maximum velocity of ∼120 µm/s and directly propagated from stimulated into neighboring endfeet; this spread was omitted in Cx30/Cx43 double-deficient mice. Sodium transients resulted in elevation of intracellular magnesium, indicating a decrease in intracellular ATP. In summary, our results establish that excitatory synaptic activity and stimulation of glutamate uptake in astrocytes trigger transient sodium increases in perivascular endfeet which rapidly spread through gap junctions into neighboring endfeet and cause a reduction of intracellular ATP. The newly discovered endfeet sodium signaling thereby represents a fast, long-lived and inter-cellularly acting indicator of synaptic activity at the blood-brain barrier, which likely constitutes an important component of neuro-metabolic coupling in the brain. GLIA 2017;65:293-308.


Assuntos
Trifosfato de Adenosina/metabolismo , Astrócitos/citologia , Junções Comunicantes/metabolismo , Ácido Glutâmico/metabolismo , Transdução de Sinais/fisiologia , Sódio/metabolismo , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Ácido Aspártico/análogos & derivados , Ácido Aspártico/farmacologia , Astrócitos/efeitos dos fármacos , Conexina 30/deficiência , Conexina 30/genética , Conexina 43/deficiência , Conexina 43/genética , Ácido D-Aspártico/farmacologia , Feminino , Junções Comunicantes/efeitos dos fármacos , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Hipocampo/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia
13.
Eur J Pharmacol ; 793: 112-118, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27838398

RESUMO

Controlling extracellular glutamate level in a physiological range is important to maintain normal sensory transmission. Here, we investigated the paradoxical action of glutamate transporters in the rat formalin test to elucidate a possible role of inversely operating transporters in its analgesic mechanism. The effects of glutamate transporter inhibitor on formalin-induced pain behavior were examined. Then we performed a microdialysis study to clarify the differential change in extracellular glutamate concentration by intrathecal administration of transportable and non-transportable blockers. And we further investigated the mechanism pharmacologically via pretreatment with antagonists of various receptors and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) staining. Intrathecally-injected glutamate transporter inhibitors, non-transportable DL-threo-ß-benzyloxyaspartat (TBOA) and transportable trans-pyrrolidine-2,4-dicarboxylic acid (t-PDC), produced paradoxical antinociception in the formalin test. In normal rats, inhibition of the glutamate transporter increased extracellular glutamate. In the formalin model rats, TBOA suppressed while t-PDC enhanced glutamate release. When tPDC was pretreated 30min prior to formalin injection, glutamate release was blocked. Blocking α-2 adrenergic receptors reversed the tPDC analgesia. Increased apoptosis was not apparent in the spinal dorsal horn of tPDC-treated rats compared to the control group. These data suggest that glutamate transporters in a formalin-induced pain state work in a reverse mode and can be blocked from releasing glutamate by TBOA and preloaded tPDC. The analgesic mechanism of TBOA may be related to the blockade of inversely operating transporter, and that of tPDC may be associated with the activation of noradrenergic neurotransmission but not with dorsal horn neurotoxicity.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Analgésicos/farmacologia , Ácido Aspártico/farmacologia , Ácidos Dicarboxílicos/farmacologia , Pirrolidinas/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Formaldeído/farmacologia , Ácido Glutâmico/metabolismo , Masculino , Nociceptividade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
14.
eNeuro ; 3(3)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27182552

RESUMO

Endocannabinoids are diffusible lipophilic molecules that may spread to neighboring synapses. Monoacylglycerol lipase (MAGL) is the principal enzyme that degrades the endocannabinoid 2-arachidonoylglycerol (2-AG). Using knock-out mice in which MAGL is deleted globally or selectively in neurons and astrocytes, we investigated the extent to which neuronal and astrocytic MAGL limit the spread of 2-AG-mediated retrograde synaptic depression in cerebellar slices. A brief tetanic stimulation of parallel fibers in the molecular layer induced synaptically evoked suppression of excitation (SSE) in Purkinje cells, and both neuronal and astrocytic MAGL contribute to the termination of this form of endocannabinoid-mediated synaptic depression. The spread of SSE among Purkinje cells occurred only after global knock-out of MAGL or pharmacological blockade of either MAGL or glutamate uptake, but no spread was detected following neuron- or astrocyte-specific deletion of MAGL. The spread of endocannabinoid signaling was also influenced by the spatial pattern of synaptic stimulation, because it did not occur at spatially dispersed parallel fiber synapses induced by stimulating the granular layer. The tetanic stimulation of parallel fibers did not induce endocannabinoid-mediated synaptic suppression in Golgi cells even after disruption of MAGL and glutamate uptake, suggesting that heightened release of 2-AG by Purkinje cells does not spread the retrograde signal to parallel fibers that innervate Golgi cells. These results suggest that both neuronal and astrocytic MAGL limit the spatial diffusion of 2-AG and confer synapse-specificity of endocannabinoid signaling.


Assuntos
Astrócitos/enzimologia , Cerebelo/enzimologia , Endocanabinoides/metabolismo , Monoacilglicerol Lipases/metabolismo , Neurônios/enzimologia , Transmissão Sináptica/fisiologia , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Ácidos Araquidônicos/farmacologia , Astrócitos/efeitos dos fármacos , Agonistas de Receptores de Canabinoides/farmacologia , Cerebelo/efeitos dos fármacos , Endocanabinoides/farmacologia , Ácido Glutâmico/metabolismo , Glicerídeos/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos Knockout , Monoacilglicerol Lipases/genética , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Transmissão Sináptica/efeitos dos fármacos , Técnicas de Cultura de Tecidos
15.
Neurochem Res ; 41(8): 2017-28, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27084771

RESUMO

Statins have been shown to promote neuroprotection in a wide range of neurological disorders. However, the mechanisms involved in such effects of statins are not fully understood. Quinolinic acid (QA) is a neurotoxin that induces seizures when infused in vivo and promotes glutamatergic excitotoxicity in the central nervous system. The aim of this study was to evaluate the putative glutamatergic mechanisms and the intracellular signaling pathways involved in the atorvastatin neuroprotective effects against QA toxicity. Atorvastatin (10 mg/kg) treatment for 7 days prevented the QA-induced decrease in glutamate uptake, but had no effect on increased glutamate release induced by QA. Moreover, atorvastatin treatment increased the phosphorylation of ERK1 and prevented the decrease in Akt phosphorylation induced by QA. Neither atorvastatin treatment nor QA infusion altered glutamine synthetase activity or the levels of phosphorylation of p38(MAPK) or JNK1/2 during the evaluation. Inhibition of MEK/ERK signaling pathway, but not PI3K/Akt signaling, abolished the neuroprotective effect of atorvastatin against QA-induced decrease in glutamate uptake. Our data suggest that atorvastatin protective effects against QA toxicity are related to modulation of glutamate transporters via MAPK/ERK signaling pathway.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Atorvastatina/farmacologia , Ácido Glutâmico/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ácido Quinolínico/toxicidade , Animais , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos
16.
Oncotarget ; 7(24): 36021-36033, 2016 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-27074570

RESUMO

The glutamate transporter xCT (SCL7a11, system Xc-, SXC) is an emerging key player in glutamate/cysteine/glutathione homeostasis in the brain and in cancer. xCT expression correlates with the grade of malignancy. Here, we report on the use of the U.S. Food and Drug Administration and EMA-approved xCT inhibitor, sulfasalazine (SAS) in gliomas. SAS does not affect cell viability in gliomas at concentrations below 200 µM. At higher concentrations SAS becomes gliomatoxic. Mechanistically SAS inhibits xCT and induces ferroptotic cell death in glioma cells. There is no evidence for impact on autophagic flux following SAS application. However, SAS can potentiate the efficacy of the standard chemotherapeutic and autophagy-inducing agent temozolomide (Temcat, Temodal or Temodar®). We also investigated SAS in non-transformed cellular constituents of the brain. Neurons and brain tissue are almost non-responding to SAS whereas isolated astrocytes are less sensitive towards SAS toxicity compared to gliomas. In vivo SAS treatment does not affect experimental tumor growth and treated animals revealed comparable tumor volume as untreated controls. However, SAS treatment resulted in reduced glioma-derived edema and, hence, total tumor volume burden as revealed by T2-weighted magnetic resonance imaging. Altogether, we show that SAS can be utilized for targeting the glutamate antiporter xCT activity as a tumor microenvironment-normalizing drug, while crucial cytotoxic effects in brain tumors are minor.


Assuntos
Edema Encefálico/prevenção & controle , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Sulfassalazina/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Animais Recém-Nascidos , Anti-Inflamatórios não Esteroides/farmacologia , Edema Encefálico/diagnóstico por imagem , Edema Encefálico/etiologia , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Glioma/complicações , Glioma/metabolismo , Humanos , Imageamento por Ressonância Magnética , Ratos Wistar , Temozolomida
17.
Calcif Tissue Int ; 99(2): 142-54, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27016923

RESUMO

Development and function of osteoblast lineage cells are regulated by a complex microenvironment consisting of the bone extracellular matrix, cells, systemic hormones and cytokines, autocrine and paracrine factors, and mechanical load. Apart from receptors that transduce extracellular signals into the cell, molecular transporters play a crucial role in the cellular response to the microenvironment. Transporter molecules are responsible for cellular uptake of nutritional components, elimination of metabolites, ion transport, and cell-cell communication. In this report, the expression of molecular transporters in osteoblast lineage cells was investigated to assess their roles in cell development and activity. Low-density arrays, covering membrane and vesicular transport molecules, were used to assess gene expression in osteoblasts representing early and late differentiation states. Receptors and transporters for the amino acid glutamate were found to be differentially expressed during osteoblast development. Glutamate is a neurotransmitter in the central nervous system, and the mechanisms of its release, signal transduction, and cellular reabsorption in the synaptic cleft are well understood. Less clear, however, is the control of equivalent processes in peripheral tissues. In primary osteoblasts, inhibition of glutamate transporters with nonselective inhibitors leads to an increase in the concentration of extracellular glutamate. This change was accompanied by a decrease in osteoblast proliferation, stimulation of alkaline phosphatase, and the expression of transcripts encoding osteocalcin. Enzymatic removal of extracellular glutamate abolished these pro-differentiation effects, as did the inhibition of PKC- and Erk1/2-signaling pathways. These findings demonstrate that glutamate signaling promotes differentiation and activation of osteoblast lineage cells. Consequently, the glutamate system may represent a putative therapeutic target to induce an anabolic response in the skeletal system. Known antagonists of glutamate transporters will serve as lead compounds in developing new and specific bioactive molecules.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Linhagem da Célula/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Osteoblastos/citologia , Receptores de Glutamato/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Osso e Ossos/metabolismo , Comunicação Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Linhagem da Célula/fisiologia , Glutamatos/metabolismo , Glutamatos/farmacologia , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteocalcina/metabolismo , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia
18.
AIDS ; 30(4): 543-52, 2016 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-26558720

RESUMO

OBJECTIVE: The neurotoxic actions of the HIV protease inhibitors, amprenavir (APV) and lopinavir (LPV) were investigated. DESIGN: With combination antiretroviral therapy (cART), HIV-infected persons exhibit neurocognitive impairments, raising the possibility that cART might exert adverse central nervous system (CNS) effects. We examined the effects of LPV and APV using in-vitro and in-vivo assays of CNS function. METHODS: Gene expression, cell viability and amino-acid levels were measured in human astrocytes, following exposure to APV or LPV. Neurobehavioral performance, amino-acid levels and neuropathology were examined in HIV-1 Vpr transgenic mice after treatment with APV or LPV. RESULTS: Excitatory amino-acid transporter-2 (EAAT2) expression was reduced in astrocytes treated with LPV or APV, especially LPV (P < 0.05), which was accompanied by reduced intracellular L-glutamate levels in LPV-treated cells (P < 0.05). Treatment of astrocytes with APV or LPV reduced the expression of proliferating cell nuclear antigen (PCNA) and Ki-67 (P < 0.05) although cell survival was unaffected. Exposure of LPV to astrocytes augmented glutamate-evoked transient rises in [Cai] (P < 0.05). Vpr mice treated with LPV showed lower concentrations of L-glutamate, L-aspartate and L-serine in cortex compared with vehicle-treated mice (P < 0.05). Total errors in T-maze assessment were increased in LPV and APV-treated animals (P < 0.05). EAAT2 expression was reduced in the brains of protease inhibitor-treated animals, which was associated with gliosis (P < 0.05). CONCLUSION: These results indicated that contemporary protease inhibitors disrupt astrocyte functions at therapeutic concentrations with enhanced sensitivity to glutamate, which can lead to neurobehavioral impairments. ART neurotoxicity should be considered in future therapeutic regimens for HIV/AIDS.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Carbamatos/efeitos adversos , Inibidores da Protease de HIV/efeitos adversos , Lopinavir/efeitos adversos , Doenças do Sistema Nervoso/induzido quimicamente , Sulfonamidas/efeitos adversos , Animais , Química Encefálica , Carbamatos/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Furanos , Perfilação da Expressão Gênica , Inibidores da Protease de HIV/administração & dosagem , HIV-1 , Humanos , Lopinavir/administração & dosagem , Masculino , Camundongos Transgênicos , Doenças do Sistema Nervoso/patologia , Exame Neurológico , Sulfonamidas/administração & dosagem
19.
Talanta ; 144: 1231-8, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26452952

RESUMO

Measurement of neurotransmitters during normal or altered function in cerebral slices could be an important tool to better understand the relationship between biochemical changes and electrophysiological activity. Some attempts of this analysis have been made; however, the current techniques do not have the appropriate time resolution to establish this relationship. The use of electrochemical biosensors has allowed for good time resolution, but problems related to the reduction of signal noise and biofouling of the electrode surface could be an important issue. In this work, we propose a new alternative to simultaneously measure glutamate and electrical activity with a high temporal resolution in brain slices. This approach is based on the use of enzymatic reactors that generate a fluorescent derivative from glutamate that can be measured at high temporal resolution. The results presented here show a reliable measurement of this neurotransmitter in brain slices obtained from intact animals under the effect of a glutamate transporter blocker DL-threo-beta-benzyloxyaspartate as well as the potassium channel blocker 4-aminopyridine. Differences in the levels of glutamate and high frequency and amplitude discharges as an effect of drug administration were found in brain slices obtained from epileptic rats (p<0.05). In conclusion, this method could be used to measure neurotransmitter concentration online at a near physiological temporal resolution, which can then be correlated to the electrical activity that is simultaneously recorded.


Assuntos
Encéfalo/citologia , Encéfalo/fisiologia , Eletrofisiologia/métodos , Espaço Extracelular/metabolismo , Ácido Glutâmico/metabolismo , 4-Aminopiridina/farmacologia , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Ácido Aspártico/farmacologia , Encéfalo/efeitos dos fármacos , Eletrofisiologia/instrumentação , Espaço Extracelular/efeitos dos fármacos , Corantes Fluorescentes/química , Ácido Glutâmico/química , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Masculino , Ratos , Ratos Wistar
20.
BMC Cancer ; 15: 411, 2015 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-25981639

RESUMO

BACKGROUND: Colorectal cancer (CRC) is a leading cause of cancer death globally and new biomarkers and treatments are severely needed. METHODS: Here, we employed HCT116 and LoVo human CRC cells made resistant to either SN38 or oxaliplatin, to investigate whether altered expression of the high affinity glutamate transporters Solute Carrier (SLC)-1A1 and -1A3 (EAAT3, EAAT1) is associated with the resistant phenotypes. Analyses included real-time quantitative PCR, immunoblotting and immunofluorescence analyses, radioactive tracer flux measurements, and biochemical analyses of cell viability and glutathione content. Results were evaluated using one- and two-way ANOVA and Students two-tailed t-test, as relevant. RESULTS: In SN38-resistant HCT116 and LoVo cells, SLC1A1 expression was down-regulated ~60 % and up-regulated ~4-fold, respectively, at both mRNA and protein level, whereas SLC1A3 protein was undetectable. The changes in SLC1A1 expression were accompanied by parallel changes in DL-Threo-ß-Benzyloxyaspartic acid (TBOA)-sensitive, UCPH101-insensitive [(3)H]-D-Aspartate uptake, consistent with increased activity of SLC1A1 (or other family members), yet not of SLC1A3. DL-TBOA co-treatment concentration-dependently augmented loss of cell viability induced by SN38, while strongly counteracting that induced by oxaliplatin, in both HCT116 and LoVo cells. This reflected neither altered expression of the oxaliplatin transporter Cu(2+)-transporter-1 (CTR1), nor changes in cellular reduced glutathione (GSH), although HCT116 cell resistance per se correlated with increased cellular GSH. DL-TBOA did not significantly alter cellular levels of p21, cleaved PARP-1, or phospho-Retinoblastoma protein, yet altered SLC1A1 subcellular localization, and reduced chemotherapy-induced p53 induction. CONCLUSIONS: SLC1A1 expression and glutamate transporter activity are altered in SN38-resistant CRC cells. Importantly, the non-selective glutamate transporter inhibitor DL-TBOA reduces chemotherapy-induced p53 induction and augments CRC cell death induced by SN38, while attenuating that induced by oxaliplatin. These findings may point to novel treatment options in treatment-resistant CRC.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Antineoplásicos/farmacologia , Ácido Aspártico/farmacologia , Camptotecina/análogos & derivados , Neoplasias Colorretais/metabolismo , Resistencia a Medicamentos Antineoplásicos , Compostos Organoplatínicos/farmacologia , Sistema X-AG de Transporte de Aminoácidos/genética , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Camptotecina/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Cobre/metabolismo , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 3 de Aminoácido Excitatório/genética , Transportador 3 de Aminoácido Excitatório/metabolismo , Expressão Gênica , Técnicas de Silenciamento de Genes , Glutationa/metabolismo , Células HCT116 , Humanos , Irinotecano , Oxaliplatina , Transporte Proteico , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA