Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Int J Mol Sci ; 22(8)2021 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-33920037

RESUMO

Epilepsy, one of the most common neurological disorders worldwide, is characterized by recurrent seizures and subsequent brain damage. Despite strong evidence supporting a deleterious impact on seizure occurrence and outcome severity, stress is an overlooked component in people with epilepsy. With regard to stressor duration and timing, acute stress can be protective in epileptogenesis, while chronic stress often promotes seizure occurrence in epilepsy patients. Preclinical research suggests that chronic stress promotes neuroinflammation and leads to a depressive state. Depression is the most common psychiatric comorbidity in people with epilepsy, resulting in a poor quality of life. Here, we summarize studies investigating acute and chronic stress as a seizure trigger and an important factor that worsens epilepsy outcomes and psychiatric comorbidities. Mechanistic insight into the impact of stress on epilepsy may create a window of opportunity for future interventions targeting neuroinflammation-related disorders.


Assuntos
Epilepsia/fisiopatologia , Inflamação/fisiopatologia , Convulsões/fisiopatologia , Comorbidade , Epilepsia/epidemiologia , Epilepsia/psicologia , Humanos , Inflamação/epidemiologia , Inflamação/psicologia , Masculino , Qualidade de Vida , Convulsões/epidemiologia , Convulsões/psicologia
2.
Int J Mol Sci ; 22(22)2021 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-34830412

RESUMO

Sleep disturbances, such as insomnia, obstructive sleep apnea, and daytime sleepiness, are common in people diagnosed with epilepsy. These disturbances can be attributed to nocturnal seizures, psychosocial factors, and/or the use of anti-epileptic drugs with sleep-modifying side effects. Epilepsy patients with poor sleep quality have intensified seizure frequency and disease progression compared to their well-rested counterparts. A better understanding of the complex relationship between sleep and epilepsy is needed, since approximately 20% of seizures and more than 90% of sudden unexpected deaths in epilepsy occur during sleep. Emerging studies suggest that neuroinflammation, (e.g., the CNS immune response characterized by the change in expression of inflammatory mediators and glial activation) may be a potential link between sleep deprivation and seizures. Here, we review the mechanisms by which sleep deprivation induces neuroinflammation and propose that neuroinflammation synergizes with seizure activity to worsen neurodegeneration in the epileptic brain. Additionally, we highlight the relevance of sleep interventions, often overlooked by physicians, to manage seizures, prevent epilepsy-related mortality, and improve quality of life.


Assuntos
Epilepsia/epidemiologia , Convulsões/epidemiologia , Privação do Sono/epidemiologia , Transtornos do Sono-Vigília/epidemiologia , Distúrbios do Sono por Sonolência Excessiva/epidemiologia , Distúrbios do Sono por Sonolência Excessiva/fisiopatologia , Epilepsia/fisiopatologia , Humanos , Doenças Neuroinflamatórias/epidemiologia , Doenças Neuroinflamatórias/fisiopatologia , Qualidade de Vida , Convulsões/fisiopatologia , Apneia Obstrutiva do Sono/epidemiologia , Apneia Obstrutiva do Sono/fisiopatologia , Privação do Sono/fisiopatologia , Distúrbios do Início e da Manutenção do Sono/epidemiologia , Distúrbios do Início e da Manutenção do Sono/fisiopatologia , Transtornos do Sono-Vigília/fisiopatologia
3.
Neurobiol Dis ; 140: 104863, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32283202

RESUMO

Seizures can be evident within minutes of exposure to an organophosphorus (OP) agent and often progress to status epilepticus (SE) resulting in a high mortality if left untreated. Effective medical countermeasures are necessary to sustain patients suffering from OP poisoning and to mitigate the ensuing brain injury. Here, the hypothesis was tested that a single subanesthetic dose of urethane prevents neuropathology measured 24 h following diisopropylfluorophosphate (DFP)-induced SE. Adult Sprague-Dawley rats were injected with DFP to induce SE. During SE rats displayed increased neuronal activity in the hippocampus and an upregulation of immediate early genes as well as pro-inflammatory mediators. In additional experiments rats were administered diazepam (10 mg/kg, ip) or urethane (0.8 g/kg, sc) 1 h after DFP-induced SE and compared to rats that experienced uninterrupted SE. Cortical electroencephalography (EEG) and power analysis strengthen the conclusion that urethane effectively terminates SE and prevents the overnight return of seizure activity. Neurodegeneration in limbic brain regions and the seizure-induced upregulation of key inflammatory mediators present 24 h after DFP-induced SE were strongly attenuated by administration of urethane. A trivial explanation for these beneficial effects, that urethane simply reactivates acetylcholinesterase, has been ruled out. These findings indicate that, by contrast to rats administered diazepam or rats that experience uninterrupted SE, the early neuropathology after SE is prevented by subanesthetic urethane, which terminates rather than interrupts, SE.


Assuntos
Isoflurofato/toxicidade , Estado Epiléptico/tratamento farmacológico , Uretana/farmacologia , Acetilcolinesterase , Animais , Lesões Encefálicas/tratamento farmacológico , Diazepam/farmacologia , Modelos Animais de Doenças , Eletroencefalografia , Inibidores Enzimáticos/farmacologia , Hipocampo/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Convulsões/tratamento farmacológico , Estado Epiléptico/induzido quimicamente
4.
Neurobiol Dis ; 133: 104399, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30818067

RESUMO

This review describes an adult rat model of status epilepticus (SE) induced by diisopropyl fluorophosphate (DFP), and the beneficial outcomes of transient inhibition of the prostaglandin-E2 receptor EP2 with a small molecule antagonist, delayed by 2-4 h after SE onset. Administration of six doses of the selective EP2 antagonist TG6-10-1 over a 2-3 day period accelerates functional recovery, attenuates hippocampal neurodegeneration, neuroinflammation, gliosis and blood-brain barrier leakage, and prevents long-term cognitive deficits without blocking SE itself or altering acute seizure characteristics. This work has provided important information regarding organophosphate-induced seizure related pathologies in adults and revealed the effectiveness of delayed EP2 inhibition to combat these pathologies.


Assuntos
Indóis/farmacologia , Intoxicação por Organofosfatos , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Estado Epiléptico/induzido quimicamente , Animais , Inibidores da Colinesterase/toxicidade , Modelos Animais de Doenças , Isoflurofato/toxicidade , Ratos
5.
Proc Natl Acad Sci U S A ; 109(8): 3149-54, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22323596

RESUMO

With interest waning in the use of cyclooxygenase-2 (COX-2) inhibitors for inflammatory disease, prostaglandin receptors provide alternative targets for the treatment of COX-2-mediated pathological conditions in both the periphery and the central nervous system. Activation of prostaglandin E2 receptor (PGE(2)) subtype EP2 promotes inflammation and is just beginning to be explored as a therapeutic target. To better understand physiological and pathological functions of the prostaglandin EP2 receptor, we developed a suite of small molecules with a 3-aryl-acrylamide scaffold as selective EP2 antagonists. The 12 most potent compounds displayed competitive antagonism of the human EP2 receptor with K(B) 2-20 nM in Schild regression analysis and 268- to 4,730-fold selectivity over the prostaglandin EP4 receptor. A brain-permeant compound completely suppressed the up-regulation of COX-2 mRNA in rat cultured microglia by EP2 activation and significantly reduced neuronal injury in hippocampus when administered in mice beginning 1 h after termination of pilocarpine-induced status epilepticus. The salutary actions of this novel group of antagonists raise the possibility that selective block of EP2 signaling via small molecules can be an innovative therapeutic strategy for inflammation-related brain injury.


Assuntos
Neurônios/efeitos dos fármacos , Neurônios/patologia , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Convulsões/metabolismo , Convulsões/patologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/biossíntese , Indução Enzimática/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/enzimologia , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Bibliotecas de Moléculas Pequenas/administração & dosagem , Bibliotecas de Moléculas Pequenas/química , Estado Epiléptico/patologia , Relação Estrutura-Atividade
6.
Neurobiol Dis ; 70: 74-89, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24952362

RESUMO

Prostaglandin E2 (PGE2) regulates membrane excitability, synaptic transmission, plasticity, and neuronal survival. The consequences of PGE2 release following seizures has been the subject of much study. Here we demonstrate that the prostaglandin E2 receptor 1 (EP1, or Ptger1) modulates native kainate receptors, a family of ionotropic glutamate receptors widely expressed throughout the central nervous system. Global ablation of the EP1 gene in mice (EP1-KO) had no effect on seizure threshold after kainate injection but reduced the likelihood to enter status epilepticus. EP1-KO mice that did experience typical status epilepticus had reduced hippocampal neurodegeneration and a blunted inflammatory response. Further studies with native prostanoid and kainate receptors in cultured cortical neurons, as well as with recombinant prostanoid and kainate receptors expressed in Xenopus oocytes, demonstrated that EP1 receptor activation potentiates heteromeric but not homomeric kainate receptors via a second messenger cascade involving phospholipase C, calcium and protein kinase C. Three critical GluK5 C-terminal serines underlie the potentiation of the GluK2/GluK5 receptor by EP1 activation. Taken together, these results indicate that EP1 receptor activation during seizures, through a protein kinase C pathway, increases the probability of kainic acid induced status epilepticus, and independently promotes hippocampal neurodegeneration and a broad inflammatory response.


Assuntos
Proteína Quinase C/metabolismo , Receptores de Ácido Caínico/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Estado Epiléptico/fisiopatologia , Animais , Células Cultivadas , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroimunomodulação/fisiologia , Oócitos/fisiologia , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP1/genética , Convulsões/fisiopatologia , Transdução de Sinais , Estado Epiléptico/patologia , Xenopus
7.
Epilepsia ; 55(1): 17-25, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24446952

RESUMO

Epilepsy is one of the more prevalent neurologic disorders in the world, affecting approximately 50 million people of different ages and backgrounds. Epileptic seizures propagating through both lobes of the forebrain can have permanent debilitating effects on a patient's cognitive and somatosensory brain functions. Epilepsy, defined by the sporadic occurrence of spontaneous recurrent seizures (SRS), is often accompanied by inflammation of the brain. Pronounced increases in the expression of key inflammatory mediators (e.g., interleukin -1ß [IL-1ß], tumor necrosis factor alpha [TNFα], cyclooxygenase-2 [COX-2], and C-X-C motif chemokine 10 [CXCL10]) after seizures may cause secondary damage in the brain and increase the likelihood of repetitive seizures. The COX-2 enzyme is induced rapidly during seizures. The increased level of COX-2 in specific areas of the epileptic brain can help to identify regions of seizure-induced brain inflammation. A good deal of effort has been expended to determine whether COX-2 inhibition might be neuroprotective and represent an adjunct therapeutic strategy along with antiepileptic drugs used to treat epilepsy. However, the effectiveness of COX-2 inhibitors on epilepsy animal models appears to depend on the timing of administration. With all of the effort placed on making use of COX-2 inhibitors as therapeutic agents for the treatment of epilepsy, inflammation, and neurodegenerative diseases there has yet to be a selective and potent COX-2 inhibitor that has shown a clear therapeutic outcome with acceptable side effects.


Assuntos
Ciclo-Oxigenase 2/fisiologia , Epilepsia/enzimologia , Animais , Anticonvulsivantes/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/enzimologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Epilepsia/tratamento farmacológico , Humanos , Inflamação/enzimologia , Doenças Neurodegenerativas/enzimologia , Convulsões/tratamento farmacológico , Convulsões/enzimologia
8.
Mol Pharmacol ; 83(4): 746-52, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23348498

RESUMO

The function of many ion channels is under dynamic control by coincident activation of G-protein-coupled receptors (GPCRs), particularly those coupled to the Gαs and Gαq family members. Such regulation is typically dependent on the subunit composition of the ionotropic receptor or channel as well as the GPCR subtype and the cell-specific panoply of signaling pathways available. Because GPCRs and ion channels are so highly represented among targets of U.S. Food and Drug Administration-approved drugs, functional cross-talk between these drug target classes is likely to underlie many therapeutic and adverse effects of marketed drugs. GPCRs engage a myriad of signaling pathways that involve protein kinases A and C (PKC) and, through PKC and interaction with ß-arrestin, Src kinase, and hence the mitogen-activated-protein-kinase cascades. We focus here on the control of ionotropic glutamate receptor function by GPCR signaling because this form of regulation can influence the strength of synaptic plasticity. The amino acid residues phosphorylated by specific kinases have been securely identified in many ionotropic glutamate (iGlu) receptor subunits, but which of these sites are GPCR targets is less well known even when the kinase has been identified. N-methyl-d-aspartate, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and heteromeric kainate receptors are all downstream targets of GPCR signaling pathways. The details of GPCR-iGlu receptor cross-talk should inform a better understanding of how synaptic transmission is regulated and lead to new therapeutic strategies for neuropsychiatric disorders.


Assuntos
Receptores Acoplados a Proteínas G/fisiologia , Receptores Ionotrópicos de Glutamato/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Transtornos Mentais/genética , Transtornos Mentais/metabolismo , Dados de Sequência Molecular , Receptor Cross-Talk/fisiologia , Receptores Acoplados a Proteínas G/química , Receptores Ionotrópicos de Glutamato/química , Receptores Ionotrópicos de Glutamato/fisiologia , Transdução de Sinais/fisiologia
9.
Mol Pharmacol ; 83(1): 106-21, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23066089

RESUMO

Kainate receptors (KARs), a family of ionotropic glutamate receptors, are widely expressed in the central nervous system and are critically involved in synaptic transmission. KAR activation is influenced by metabotropic glutamate receptor (mGlu) signaling, but the underlying mechanisms are not understood. We undertook studies to examine how mGlu modulation affects activation of KARs. Confocal immunohistochemistry of rat hippocampus and cultured rat cortex revealed colocalization of the high-affinity KAR subunits with group I mGlu receptors. In hippocampal and cortical cultures, the calcium signal caused by activation of native KARs was potentiated by activation of group I mGlu receptors. In Xenopus laevis oocytes, activation of group I mGlu receptors potentiated heteromeric but not homomeric KAR-mediated currents, with no change in agonist potency. The potentiation of heteromeric KARs by mGlu1 activation was attenuated by GDPßS, blocked by an inhibitor of phospholipase C or the calcium chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA), prolonged by the phosphatase inhibitor okadaic acid, but unaffected by the tyrosine kinase inhibitor lavendustin A. Protein kinase C (PKC) inhibition reduced the potentiation by mGlu1 of GluK2/GluK5, and conversely, direct activation of PKC by phorbol 12-myristate,13-acetate potentiated GluK2/GluK5. Using site-directed mutagenesis, we identified three serines (Ser833, Ser836, and Ser840) within the membrane proximal region of the GluK5 C-terminal domain that, in combination, are required for mGlu1-mediated potentiation of KARs. Together, these data suggest that phosphorylation of key residues in the C-terminal domain changes the overall charge of this domain, resulting in potentiated agonist responses.


Assuntos
Encéfalo/metabolismo , Receptores de Ácido Caínico/fisiologia , Receptores de Glutamato Metabotrópico/agonistas , Animais , Encéfalo/citologia , Cálcio/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Ativação Enzimática , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Masculino , Oócitos/fisiologia , Técnicas de Patch-Clamp , Fosforilação , Proteína Quinase C/metabolismo , Multimerização Proteica , Subunidades Proteicas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Ácido Caínico/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Fosfolipases Tipo C/metabolismo , Xenopus laevis
10.
Proc Natl Acad Sci U S A ; 107(5): 2307-12, 2010 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20080612

RESUMO

Activation of the Galphas-coupled EP2 receptor for prostaglandin E2 (PGE(2)) promotes cell survival in several models of tissue damage. To advance understanding of EP2 functions, we designed experiments to develop allosteric potentiators of this key prostaglandin receptor. Screens of 292,000 compounds identified 93 that at 20 microM (i) potentiated the cAMP response to a low concentration of PGE(2) by > 50%; (ii) had no effect on EP4 or beta2 adrenergic receptors, the cAMP assay itself, or the parent cell line; and (iii) increased the potency of PGE(2) on EP2 receptors at least 3-fold. In aqueous solution, the active compounds are largely present as nanoparticles that appear to serve as active reservoirs for bioactive monomer. From 94 compounds synthesized or purchased, based on the modification of one hit compound, the most active increased the potency of PGE(2) on EP2 receptors 4- to 5-fold at 10 to 20 microM and showed substantial neuroprotection in an excitotoxicity model. These small molecules represent previously undescribed allosteric modulators of a PGE(2) receptor. Our results strongly reinforce the notion that activation of EP2 receptors by endogenous PGE(2) released in a cell-injury setting is neuroprotective.


Assuntos
Fármacos Neuroprotetores/farmacologia , Receptores de Prostaglandina E/agonistas , Regulação Alostérica , Animais , Técnicas Biossensoriais , AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Avaliação Pré-Clínica de Medicamentos , Transferência Ressonante de Energia de Fluorescência , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Técnicas In Vitro , Nanopartículas , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/química , Ratos , Ratos Endogâmicos SHR , Receptores de Prostaglandina E Subtipo EP2 , Relação Estrutura-Atividade
11.
Neurotoxicology ; 91: 45-59, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35500718

RESUMO

Exposure to high levels of a cholinesterase inhibiting organophosphorus (OP) agent often results in seizures that progress to status epilepticus (SE). Survivors of OP-induced SE often display neuropathological consequences the days following SE. In the current study, the temporal profile of neuropathology after SE was investigated in a rat model of diisopropylfluorophosphate (DFP)-induced SE. Adult Sprague-Dawley rats were injected with DFP to induce SE for one hour. Following termination of electrographic SE with urethane (0.8 g/kg, sc), cohorts of rats were euthanized 3, 24 and 48 h later and brain tissue was processed to determine immediate early gene and inflammatory mediator expression as well as blood-brain barrier changes and neurodegeneration. After SE rats displayed a time-dependent upregulation of immediate early genes such as cFos and ΔFosB as well as pro-inflammatory mediators COX-2, IL-1ß and IL-6. The profile of positive cFos staining, but not ΔFosB, coincided temporally with heightened brain activity measured by cortical electroencephalography (EEG). Neurodegeneration in limbic brain regions was absent 3 h after SE, but prominent 24 h later and continued to increase 48 h after SE. Serum albumin was detected in the cortex 3 h after SE suggesting early loss of blood brain barrier integrity. However, the blood-brain barrier appeared repaired 48 h after SE. This study demonstrates that following OP-poisoning in rats, immediate early gene expression in the brain precedes neuroinflammation followed by erosion of the blood-brain barrier and neurodegeneration. The study also demonstrates that seizure activity in brain nuclei coincides with cFos expression. Together, these studies give insight into the temporal molecular changes in the brain following organophosphate-induced status epilepticus.


Assuntos
Intoxicação por Organofosfatos , Estado Epiléptico , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Isoflurofato/toxicidade , Intoxicação por Organofosfatos/metabolismo , Organofosfatos/efeitos adversos , Ratos , Ratos Sprague-Dawley , Estado Epiléptico/patologia
12.
Biomed Pharmacother ; 147: 112646, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35091236

RESUMO

The EP2 receptor has emerged as a therapeutic target with exacerbating role in disease pathology for a variety of peripheral and central nervous system disorders. We and others have recently demonstrated beneficial effects of EP2 antagonists in preclinical models of neuroinflammation and peripheral inflammation. However, it was earlier reported that mice with global EP2 knockout (KO) display adverse phenotypes on fertility and blood pressure. Other studies indicated that EP2 activation with an agonist has a beneficial effect of healing fractured bone in animal models. These results impeded the development of EP2 antagonists, and EP2 antagonism as therapeutic strategy. To determine whether treatment with EP2 antagonist mimics the adverse phenotypes of the EP2 global KO mouse, we tested two EP2 antagonists TG11-77. HCl and TG6-10-1 in mice and rats while they are on normal or high-salt diet, and by two different administration protocols (acute and chronic). There were no adverse effects of the antagonists on systolic and diastolic blood pressure, heart rate, respiratory function in mice and rats regardless of rodents being on a regular or high salt diet. Furthermore, chronic exposure to TG11-77. HCl produced no adverse effects on blood cell counts, bone-volume and bone-mineral density in mice. Our findings argue against adverse effects on cardiovascular and respiratory systems, blood counts and bone structure in healthy rodents from the use of small molecule reversible antagonists for EP2, in contrast to the genetic ablation model. This study paves the way for advancing therapeutic applications of EP2 antagonists against diseases involving EP2 dysfunction.


Assuntos
Doenças Cardiovasculares/patologia , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Animais , Contagem de Células Sanguíneas , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Hemodinâmica/efeitos dos fármacos , Indóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Ratos , Ratos Sprague-Dawley , Taxa Respiratória/efeitos dos fármacos
13.
Neurotoxicology ; 83: 14-27, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33352274

RESUMO

The increasing number of cases involving the use of nerve agents as deadly weapons has spurred investigation into the molecular mechanisms underlying nerve agent-induced pathology. The highly toxic nature of nerve agents restrict their use in academic research laboratories. Less toxic organophosphorus (OP) based agents including diisopropylfluorophosphate (DFP) are used as surrogates in academic research laboratories to mimic nerve agent poisoning. However, neuropathology resulting from DFP-induced status epilepticus (SE) has not been compared directly to neuropathology observed following nerve agent poisoning in the same study. Here, the hypothesis that neuropathology measured four days after SE is the same for rats exposed to DFP and soman was tested. Adult Sprague-Dawley rats were injected with soman or DFP to induce SE. Cortical electroencephalography (EEG) was recorded prior to and during soman-induced SE. EEG power analysis of rats administered soman revealed prolonged electrographic SE similar to that of rats that endure uninterrupted SE following injection of DFP. Rats that experienced soman-induced SE displayed less hippocampal neuroinflammation and gliosis compared to rats administered DFP. Seizure-induced weight change, blood-brain barrier (BBB) leakiness and neurodegeneration in most seizure sensitive limbic brain regions were similar for rats that endured SE following soman or DFP. The amalgamated pathology score calculated by combining pathological measures (weight loss, hippocampal neuroinflammation, gliosis, BBB integrity and neurodegeneration) was similar in rats administered the OP agents. These findings support use of the rat DFP model of SE as a suitable surrogate for investigating some, but not all delayed consequences produced by nerve agents.


Assuntos
Encéfalo/patologia , Encefalite/patologia , Isoflurofato , Soman , Estado Epiléptico/patologia , Animais , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Ondas Encefálicas , Morte Celular , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Eletroencefalografia , Encefalite/induzido quimicamente , Encefalite/metabolismo , Encefalite/fisiopatologia , Gliose , Masculino , Ratos Sprague-Dawley , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/metabolismo , Estado Epiléptico/fisiopatologia , Fatores de Tempo , Redução de Peso
14.
Neurotherapeutics ; 18(2): 1207-1225, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33410110

RESUMO

Prostaglandin-E2 (PGE2), an important mediator of inflammation, achieves its functions via four different G protein-coupled receptors (EP1, EP2, EP3, and EP4). We previously demonstrated that the EP2 receptor plays a proinflammatory and neurodegenerative role after status epilepticus (SE). We recently developed TG8-260 as a second-generation highly potent and selective EP2 antagonist. Here, we investigate whether TG8-260 is anti-inflammatory and combats neuropathology caused by pilocarpine-induced SE in rats. Adult male Sprague-Dawley rats were injected subcutaneously with pilocarpine (380-400 mg/kg) to induce SE. Following 60 min of SE, the rats were administered three doses of TG8-260 or vehicle and were allowed to recover. Neurodegeneration, neuroinflammation, gliosis, and blood-brain barrier (BBB) integrity were examined 4 days after SE. The results confirmed that pilocarpine-induced SE results in hippocampal neurodegeneration and a robust inflammatory response that persists days after SE. Furthermore, inhibition of the EP2 receptor by TG8-260 administered beginning 2 h after SE significantly reduced hippocampal neuroinflammation and gliosis but, in distinction to the earlier generation EP2 antagonist, did not mitigate neuronal injury or BBB breakdown. Thus, attenuation of neuroinflammation and gliosis is a common feature of EP2 inhibition following SE.


Assuntos
Gliose/tratamento farmacológico , Mediadores da Inflamação/antagonistas & inibidores , Antagonistas de Prostaglandina/uso terapêutico , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Estado Epiléptico/tratamento farmacológico , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Gliose/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Pilocarpina/toxicidade , Antagonistas de Prostaglandina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/metabolismo
15.
J Physiol ; 588(Pt 4): 683-700, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20026616

RESUMO

Kainate receptor subunits can form functional channels as homomers of GluK1, GluK2 or GluK3, or as heteromeric combinations with each other or incorporating GluK4 or GluK5 subunits. However, GluK4 and GluK5 cannot form functional channels by themselves. Incorporation of GluK4 or GluK5 into a heteromeric complex increases glutamate apparent affinity and also enables receptor activation by the agonist AMPA. Utilizing two-electrode voltage clamp of Xenopus oocytes injected with cRNA encoding kainate receptor subunits, we have observed that heteromeric channels composed of GluK2/GluK4 and GluK2/GluK5 have steady state concentration-response curves that were bell-shaped in response to either glutamate or AMPA. By contrast, homomeric GluK2 channels exhibited a monophasic steady state concentration-response curve that simply plateaued at high glutamate concentrations. By fitting several specific Markov models to GluK2/GluK4 heteromeric and GluK2 homomeric concentration-response data, we have determined that: (a) two strikingly different agonist binding affinities exist; (b) the high-affinity binding site leads to channel opening; and (c) the low-affinity agonist binding site leads to strong desensitization after agonist binding. Model parameters also approximate the onset and recovery kinetics of desensitization observed for macroscopic currents measured from HEK-293 cells expressing GluK2 and GluK4 subunits. The GluK2(E738D) mutation lowers the steady state apparent affinity for glutamate by 9000-fold in comparison to GluK2 homomeric wildtype receptors. When this mutant subunit was expressed with GluK4, the rising phase of the glutamate steady state concentration-response curve overlapped with the wildtype curve, whereas the declining phase was right-shifted toward lower affinity. Taken together, these data are consistent with a scheme whereby high-affinity agonist binding to a non-desensitizing GluK4 subunit opens the heteromeric channel, whereas low-affinity agonist binding to GluK2 desensitizes the whole channel complex.


Assuntos
Receptores de Ácido Caínico/fisiologia , Animais , Sítios de Ligação/fisiologia , Ácido Glutâmico/fisiologia , Cadeias de Markov , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Receptores de Ácido Caínico/genética , Xenopus laevis
16.
Biochem Biophys Res Commun ; 394(2): 285-90, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20193660

RESUMO

Rett syndrome is a neurodevelopmental disorder caused by Mecp2 gene mutations. In RTT patients and Mecp2-null (Mecp2(-/Y)) mice, norepinephrine (NE) content drops significantly, which may play a role in breathing arrhythmia, sleep disorders and sudden death. However, the underlying mechanisms for the NE defect are not fully understood. The NE defect may result from decreased NE biosynthesis, loss of catecholaminergic neurons or both. Although deficiency in tyrosine hydroxylase (TH) has been demonstrated, it is possible that dopamine beta-hydroxylase (DBH), the critical enzyme converting dopamine to NE, is also affected. To test these possibilities, we studied DBH expressions in pontine catecholaminergic neurons of Mecp2(-/Y) mice identified with breathing abnormalities. In comparison to the wild type, Mecp2(-/Y) mice at 2months of age showed approximately 50% decrease in the expressions of DBH and TH, at both protein and mRNA levels in the locus coeruleus (LC) region. Consistently, DBH and TH immunoreactivity was markedly decreased in LC neurons of Mecp2(-/Y) mice. No evidence was found for selective deficiency in TH- or DBH-containing neurons in Mecp2(-/Y) mice, as almost all TH-positive cells expressed DBH. By counting TH-immunoreactive cells in the LC, we found that the Mecp2(-/Y) mice lost only approximately 5% of the catecholaminergic neurons as compared to wild-type, although their LC volume shrank by approximately 15%. These results strongly suggest that the NE defect in Mecp2(-/Y) mice is likely to result from deficient expression of not only TH but also DBH without significant loss of catecholaminergic neurons in the LC.


Assuntos
Dopamina beta-Hidroxilase/deficiência , Proteína 2 de Ligação a Metil-CpG/genética , Neurônios/enzimologia , Norepinefrina/deficiência , Ponte/enzimologia , Síndrome de Rett/enzimologia , Animais , Catecolaminas/metabolismo , Dopamina beta-Hidroxilase/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Norepinefrina/biossíntese , Anormalidades do Sistema Respiratório/enzimologia , Anormalidades do Sistema Respiratório/genética , Síndrome de Rett/genética , Transcrição Gênica , Tirosina 3-Mono-Oxigenase/deficiência
17.
J Med Chem ; 63(3): 1032-1050, 2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-31904232

RESUMO

Activation of prostanoid EP2 receptor exacerbates neuroinflammatory and neurodegenerative pathology in central nervous system diseases such as epilepsy, Alzheimer's disease, and cerebral aneurysms. A selective and brain-permeable EP2 antagonist will be useful to attenuate the inflammatory consequences of EP2 activation and to reduce the severity of these chronic diseases. We recently developed a brain-permeable EP2 antagonist 1 (TG6-10-1), which displayed anti-inflammatory and neuroprotective actions in rodent models of status epilepticus. However, this compound exhibited moderate selectivity to EP2, a short plasma half-life in rodents (1.7 h) and low aqueous solubility (27 µM), limiting its use in animal models of chronic disease. With lead-optimization studies, we have developed several novel EP2 antagonists with improved water solubility, brain penetration, high EP2 potency, and selectivity. These novel inhibitors suppress inflammatory gene expression induced by EP2 receptor activation in a microglial cell line, reinforcing the use of EP2 antagonists as anti-inflammatory agents.


Assuntos
Anti-Inflamatórios/farmacologia , Indóis/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Animais , Anti-Inflamatórios/síntese química , Anti-Inflamatórios/farmacocinética , Linhagem Celular , Doenças do Sistema Nervoso Central/metabolismo , Humanos , Indóis/síntese química , Indóis/farmacocinética , Masculino , Camundongos , Microglia/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/farmacocinética , Solubilidade , Relação Estrutura-Atividade , Água/química
18.
ACS Chem Neurosci ; 10(10): 4280-4292, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31469538

RESUMO

Recently, EP2 signaling pathways were shown to regulate the classical activation and death of microglia in rat primary microglial culture. The study of microglial cells has been challenging because they are time-consuming to isolate in culture, they are demanding in their growth requirements, and they have a limited lifespan. To circumvent these difficulties, we created a murine BV2 microglial cell line stably expressing human EP2 receptors (BV2-hEP2) and further explored EP2 modulation of microglial functions. The BV2-hEP2 cells displayed cAMP elevation when exposed to the selective EP2 receptor agonists (ONO-AE1-259-1 and CP544326), and this response was competitively inhibited by TG4-155, a selective EP2 antagonist (Schild KB = 2.6 nM). By contrast, untransfected BV2 cells were unresponsive to selective EP2 agonists. Similar to the case of rat primary microglia, BV2-hEP2 microglia treated with lipopolysaccharide (LPS) (100 ng/mL) displayed rapid and robust induction of the inflammatory mediators COX-2, IL-1ß, TNFα, and IL-6. EP2 activation depressed TNFα induction but exacerbated that of the other inflammatory mediators. Like primary microglia, classically activated BV2 microglia phagocytose fluorescent-labeled latex microspheres. The presence of EP2, but not its activation by agonists, in BV2-hEP2 microglia reduced phagocytosis and proliferation by 65% and 32%, respectively, compared to BV2 microglia. Thus, BV2-hEP2 is the first microglial cell line that retains the EP2 modulation of immune regulation and phagocytic ability of native microglia. Suppression of phagocytosis by the EP2 protein appears unrelated to classical EP2 signaling pathways, which has implications for therapeutic development of EP2 antagonists.


Assuntos
Microglia/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Acetatos/farmacologia , Animais , Linhagem Celular , Proliferação de Células/fisiologia , AMP Cíclico/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/farmacologia , Humanos , Camundongos , Microglia/efeitos dos fármacos , Fagocitose/fisiologia , Receptores de Prostaglandina E Subtipo EP2/genética , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
19.
Expert Opin Ther Targets ; 23(1): 1-13, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30484341

RESUMO

Introduction:A robust neuroinflammatory response is a prevalent feature of multiple neurological disorders, including epilepsy and acute status epilepticus. One component of this neuroinflammatory reaction is the induction of cyclooxygenase-2 (COX-2), synthesis of several prostaglandins and endocannabinoid metabolites, and subsequent activation of prostaglandin and related receptors. Neuroinflammation mediated by COX-2 and its downstream effectors has received considerable attention as a potential target class to ameliorate the deleterious consequences of neurological injury. Areas covered: Here we describe the roles of COX-2 as a major inflammatory mediator. In addition, we discuss the receptors for prostanoids PGE2, prostaglandin D2, and PGF2α as potential therapeutic targets for inflammation-driven diseases. The consequences of prostanoid receptor activation after seizure activity are discussed with an emphasis on the utilization of small molecules to modulate prostanoid receptor activity. Expert opinion: Limited clinical trial experience is supportive but not definitive for a role of the COX signaling cascade in epileptogenesis. The cardiotoxicity associated with chronic coxib use, and the expectation that COX-2 inhibition will influence the levels of endocannabinoids, leukotrienes, and lipoxins as well as the prostaglandins and their endocannabinoid metabolite analogs, is shifting attention toward downstream synthases and receptors that mediate inflammation in the brain.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Epilepsia/fisiopatologia , Terapia de Alvo Molecular , Animais , Encéfalo/fisiopatologia , Ciclo-Oxigenase 2/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/efeitos adversos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Epilepsia/tratamento farmacológico , Humanos , Inflamação/fisiopatologia , Prostaglandinas/metabolismo , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina E/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
J Alzheimers Dis ; 70(4): 1259-1274, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31322556

RESUMO

Alzheimer's disease (AD) pathology consists of extracellular deposits of amyloid-ß peptides (Aß) and intracellular neurofibrillary tangles. These pathological alterations are accompanied by a neuroinflammatory response consisting of increased expression of inflammatory mediators. An anti-inflammatory strategy designed to prevent or delay the development of AD would benefit from knowing when neuroinflammation appears in the transgenic models during prodromal disease stages relative to Aß pathology. We investigated the expression patterns of inflammatory mediators in the brain of 5xFAD mice in comparison to development of Aß deposition. Expression changes in inflammatory mediators and glial markers are more robust in female mice starting at three months of age, in contrast to males in which there is no clear trend through five months. Female and male 5xFAD mice also displayed an age-dependent increase in cortical Aß deposition congruent with neuroinflammation. Thus, in the 5xFAD mouse model of AD, administration of an anti-inflammatory agent would be most efficacious when administered before three months of age.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Mediadores da Inflamação/metabolismo , Sintomas Prodrômicos , Caracteres Sexuais , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA