Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(36): e2321874121, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39207736

RESUMO

Medium chain fatty acids are commonly consumed as part of diets for endurance sports and as medical treatment in ketogenic diets where these diets regulate energy metabolism and increase adenosine levels. However, the role of the equilibrative nucleoside transporter 1 (ENT1), which is responsible for adenosine transport across membranes in this process, is not well understood. Here, we investigate ENT1 activity in controlling the effects of two dietary medium chain fatty acids (decanoic and octanoic acid), employing the tractable model system Dictyostelium. We show that genetic ablation of three ENT1 orthologues unexpectedly improves cell proliferation specifically following decanoic acid treatment. This effect is not caused by increased adenosine levels triggered by both fatty acids in the presence of ENT1 activity. Instead, we show that decanoic acid increases expression of energy-related genes relevant for fatty acid ß-oxidation, and that pharmacological inhibition of ENT1 activity leads to an enhanced effect of decanoic acid to increase expression of tricarboxylicacid cycle and oxidative phosphorylation components. Importantly, similar transcriptional changes have been shown in the rat hippocampus during ketogenic diet treatment. We validated these changes by showing enhanced mitochondria load and reduced lipid droplets. Thus, our data show that ENT1 regulates the medium chain fatty acid-induced increase in cellular adenosine levels and the decanoic acid-induced expression of important metabolic enzymes in energy provision, identifying a key role for ENT1 proteins in metabolic effects of medium chain fatty acids.


Assuntos
Metabolismo Energético , Transportador Equilibrativo 1 de Nucleosídeo , Adenosina/metabolismo , Adenosina/farmacologia , Caprilatos/farmacologia , Proliferação de Células/efeitos dos fármacos , Dictyostelium/metabolismo , Dictyostelium/genética , Dictyostelium/efeitos dos fármacos , Dieta Cetogênica , Gorduras na Dieta/farmacologia , Gorduras na Dieta/metabolismo , Metabolismo Energético/efeitos dos fármacos , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Transportador Equilibrativo 1 de Nucleosídeo/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos
2.
Int J Mol Sci ; 25(12)2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38928292

RESUMO

Tanshinone IIA (T2A) is a bioactive compound that provides promise in the treatment of glioblastoma multiforme (GBM), with a range of molecular mechanisms including the inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) and the induction of autophagy. Recently, T2A has been demonstrated to function through sestrin 2 (SESN) to inhibit mTORC1 activity, but its possible impact on autophagy through this pathway has not been investigated. Here, the model system Dictyostelium discoideum and GBM cell lines were employed to investigate the cellular role of T2A in regulating SESN to inhibit mTORC1 and activate autophagy through a GATOR2 component MIOS. In D. discoideum, T2A treatment induced autophagy and inhibited mTORC1 activity, with both effects lost upon the ablation of SESN (sesn-) or MIOS (mios-). We further investigated the targeting of MIOS to reproduce this effect of T2A, where computational analysis identified 25 novel compounds predicted to strongly bind the human MIOS protein, with one compound (MIOS inhibitor 3; Mi3) reducing cell proliferation in two GBM cells. Furthermore, Mi3 specificity was demonstrated through the loss of potency in the D. discoideum mios- cells regarding cell proliferation and the induction of autophagy. In GBM cells, Mi3 treatment also reduced mTORC1 activity and induced autophagy. Thus, a potential T2A mimetic showing the inhibition of mTORC1 and induction of autophagy in GBM cells was identified.


Assuntos
Abietanos , Autofagia , Dictyostelium , Glioblastoma , Alvo Mecanístico do Complexo 1 de Rapamicina , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Abietanos/farmacologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Dictyostelium/efeitos dos fármacos , Dictyostelium/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/antagonistas & inibidores , Sestrinas
3.
Plant Commun ; 5(6): 100846, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38460510

RESUMO

Allelochemicals represent a class of natural products released by plants as root, leaf, and fruit exudates that interfere with the growth and survival of neighboring plants. Understanding how allelochemicals function to regulate plant responses may provide valuable new approaches to better control plant function. One such allelochemical, Myrigalone A (MyA) produced by Myrica gale, inhibits seed germination and seedling growth through an unknown mechanism. Here, we investigate MyA using the tractable model Dictyostelium discoideum and reveal that its activity depends on the conserved homolog of the plant ethylene synthesis protein 1-aminocyclopropane-1-carboxylic acid oxidase (ACO). Furthermore, in silico modeling predicts the direct binding of MyA to ACO within the catalytic pocket. In D. discoideum, ablation of ACO mimics the MyA-dependent developmental delay, which is partially restored by exogenous ethylene, and MyA reduces ethylene production. In Arabidopsis thaliana, MyA treatment delays seed germination, and this effect is rescued by exogenous ethylene. It also mimics the effect of established ACO inhibitors on root and hypocotyl extension, blocks ethylene-dependent root hair production, and reduces ethylene production. Finally, in silico binding analyses identify a range of highly potent ethylene inhibitors that block ethylene-dependent response and reduce ethylene production in Arabidopsis. Thus, we demonstrate a molecular mechanism by which the allelochemical MyA reduces ethylene biosynthesis and identify a range of ultrapotent inhibitors of ethylene-regulated responses.


Assuntos
Arabidopsis , Etilenos , Feromônios , Etilenos/biossíntese , Etilenos/metabolismo , Feromônios/farmacologia , Feromônios/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Arabidopsis/efeitos dos fármacos , Germinação/efeitos dos fármacos
4.
Cell Death Discov ; 9(1): 172, 2023 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-37202382

RESUMO

Glioblastomas are a highly aggressive cancer type which respond poorly to current pharmaceutical treatments, thus novel therapeutic approaches need to be investigated. One such approach involves the use of the bioactive natural product Tanshinone IIA (T2A) derived from the Chinese herb Danshen, where mechanistic insight for this anti-cancer agent is needed to validate its use. Here, we employ a tractable model system, Dictyostelium discoideum, to provide this insight. T2A potently inhibits cellular proliferation of Dictyostelium, suggesting molecular targets in this model. We show that T2A rapidly reduces phosphoinositide 3 kinase (PI3K) and protein kinase B (PKB) activity, but surprisingly, the downstream complex mechanistic target of rapamycin complex 1 (mTORC1) is only inhibited following chronic treatment. Investigating regulators of mTORC1, including PKB, tuberous sclerosis complex (TSC), and AMP-activated protein kinase (AMPK), suggests these enzymes were not responsible for this effect, implicating an additional molecular mechanism of T2A. We identify this mechanism as the increased expression of sestrin, a negative regulator of mTORC1. We further show that combinatory treatment using a PI3K inhibitor and T2A gives rise to a synergistic inhibition of cell proliferation. We then translate our findings to human and mouse-derived glioblastoma cell lines, where both a PI3K inhibitor (Paxalisib) and T2A reduces glioblastoma proliferation in monolayer cultures and in spheroid expansion, with combinatory treatment significantly enhancing this effect. Thus, we propose a new approach for cancer treatment, including glioblastomas, through combinatory treatment with PI3K inhibitors and T2A.

5.
Artigo em Inglês | MEDLINE | ID: mdl-36568268

RESUMO

The rationale for using thalidomide (THD) as a treatment for nausea and vomiting during pregnancy in the late 1950s appears to have been based on its sedative or hypnotic properties. In contrast to contemporaneous studies on the anti-emetic activity of phenothiazines, we were unable to identify publications reporting preclinical or clinical evaluation of THD as an anti-emetic. Our survey of the literature revealed a clinical study in 1965 showing THD reduced vomiting in cancer chemotherapy which was substantiated by similar studies from 2000, particularly showing efficacy in the delayed phase of chemotherapy-induced nausea and vomiting. To identify the mechanism(s) potentially involved in thalidomide's anti-emetic activity we reviewed its pharmacology in the light of nausea and vomiting mechanisms and their pharmacology with a particular emphasis on chemotherapy and pregnancy. The process identified the following potential mechanisms: reduced secretion of Growth Differentiation Factor 15, suppression of inflammation/prostaglandin production, downregulation of cytotoxic drug induced upregulation of iNOS, and modulation of BK (KCa1.1) channels and GABAA/glutamate transmission at critical points in the emetic pathways (nucleus tractus solitarius, area postrema). We propose ways to investigate these hypothesized mechanisms and discuss the associated challenges (e.g., objective quantification of nausea) in addition to some of the more general aspects of developing novel drugs to treat nausea and vomiting.

6.
Br J Pharmacol ; 179(14): 3628-3644, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-32959886

RESUMO

BACKGROUND AND PURPOSE: AMPA receptors, which shape excitatory postsynaptic currents and are directly involved in overactivation of synaptic function during seizures, represent a well-accepted target for anti-epileptic drugs. Trans-4-butylcyclohexane carboxylic acid (4-BCCA) has emerged as a new promising anti-epileptic drug in several in vitro and in vivo seizure models, but the mechanism of its action remained unknown. The purpose of this study is to characterize structure and dynamics of 4-BCCA interaction with AMPA receptors. EXPERIMENTAL APPROACH: We studied the molecular mechanism of AMPA receptor inhibition by 4-BCCA using a combination of X-ray crystallography, mutagenesis, electrophysiological assays, and molecular dynamics simulations. KEY RESULTS: We identified 4-BCCA binding sites in the transmembrane domain (TMD) of AMPA receptor, at the lateral portals formed by transmembrane segments M1-M4. At this binding site, 4-BCCA is very dynamic, assumes multiple poses, and can enter the ion channel pore. CONCLUSION AND IMPLICATIONS: 4-BCCA represents a low-affinity inhibitor of AMPA receptors that acts at the TMD sites distinct from non-competitive inhibitors, such as the anti-epileptic drug perampanel and the ion channel blockers. Further studies might examine the possibsility of synergistic use of these inhibitors in treatment of epilepsy and a wide range of neurological disorders and gliomas. LINKED ARTICLES: This article is part of a themed issue on Structure Guided Pharmacology of Membrane Proteins (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.14/issuetoc.


Assuntos
Ácidos Carboxílicos , Receptores de AMPA , Cicloexanos , Humanos , Receptores de AMPA/metabolismo , Convulsões
7.
Cells ; 10(11)2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34831171

RESUMO

Ketogenic diets, used in epilepsy treatment, are considered to work through reduced glucose and ketone generation to regulate a range of cellular process including autophagy induction. Recent studies into the medium-chain triglyceride (MCT) ketogenic diet have suggested that medium-chain fatty acids (MCFAs) provided in the diet, decanoic acid and octanoic acid, cause specific therapeutic effects independent of glucose reduction, although a role in autophagy has not been investigated. Both autophagy and MCFAs have been widely studied in Dictyostelium, with findings providing important advances in the study of autophagy-related pathologies such as neurodegenerative diseases. Here, we utilize this model to analyze a role for MCFAs in regulating autophagy. We show that treatment with decanoic acid but not octanoic acid induces autophagosome formation and modulates autophagic flux in high glucose conditions. To investigate this effect, decanoic acid, but not octanoic acid, was found to induce the expression of autophagy-inducing proteins (Atg1 and Atg8), providing a mechanism for this effect. Finally, we demonstrate a range of related fatty acid derivatives with seizure control activity, 4BCCA, 4EOA, and Epilim (valproic acid), also function to induce autophagosome formation in this model. Thus, our data suggest that decanoic acid and related compounds may provide a less-restrictive therapeutic approach to activate autophagy.


Assuntos
Autofagia , Ácidos Decanoicos/farmacologia , Dictyostelium/citologia , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagia/efeitos dos fármacos , Dictyostelium/efeitos dos fármacos , Dictyostelium/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Front Cell Dev Biol ; 9: 722066, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34589488

RESUMO

Ketogenic diets have been utilized for many years to improve health, and as a dietary approach for the treatment of a range of diseases, where the mechanism of these low carbohydrate and high fat diets is widely considered to be through the production of metabolic products of fat breakdown, called ketones. One of these diets, the medium chain triglyceride ketogenic diet, involves high fat dietary intake in the form of medium chain fatty acids (MCFAs), decanoic and octanoic acid, and is commonly used in endurance and high intensity exercises but has also demonstrated beneficial effects in the treatment of numerous pathologies including drug resistant epilepsy, cancer, and diabetes. Recent advances, using Dictyostelium discoideum as a model, have controversially proposed several direct molecular mechanisms for decanoic acid in this diet, independent of ketone generation. Studies in this model have identified that decanoic acid reduces phosphoinositide turnover, diacylglycerol kinase (DGK) activity, and also inhibits the mechanistic target of rapamycin complex 1 (mTORC1). These discoveries could potentially impact the treatment of a range of disorders including epilepsy, cancer and bipolar disorder. In this review, we summarize the newly proposed mechanisms for decanoic acid, identified using D. discoideum, and highlight potential roles in health and disease treatment.

9.
Br J Pharmacol ; 178(5): 1149-1163, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33347604

RESUMO

BACKGROUND AND PURPOSE: Cannabidiol (CBD) has been shown to differentially regulate the mechanistic target of rapamycin complex 1 (mTORC1) in preclinical models of disease, where it reduces activity in models of epilepsies and cancer and increases it in models of multiple sclerosis (MS) and psychosis. Here, we investigate the effects of phytocannabinoids on mTORC1 and define a molecular mechanism. EXPERIMENTAL APPROACH: A novel mechanism for phytocannabinoids was identified using the tractable model system, Dictyostelium discoideum. Using mouse embryonic fibroblasts, we further validate this new mechanism of action. We demonstrate clinical relevance using cells derived from healthy individuals and from people with MS (pwMS). KEY RESULTS: Both CBD and the more abundant cannabigerol (CBG) enhance mTORC1 activity in D. discoideum. We identify a mechanism for this effect involving inositol polyphosphate multikinase (IPMK), where elevated IPMK expression reverses the response to phytocannabinoids, decreasing mTORC1 activity upon treatment, providing new insight on phytocannabinoids' actions. We further validated this mechanism using mouse embryonic fibroblasts. Clinical relevance of this effect was shown in primary human peripheral blood mononuclear cells, where CBD and CBG treatment increased mTORC1 activity in cells derived from healthy individuals and decreased mTORC1 activity in cells derived from pwMS. CONCLUSION AND IMPLICATIONS: Our findings suggest that both CBD and the abundant CBG differentially regulate mTORC1 signalling through a mechanism dependent on the activity of the upstream IPMK signalling pathway, with potential relevance to the treatment of mTOR-related disorders, including MS.


Assuntos
Canabinoides/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Fosfotransferases (Aceptor do Grupo Álcool) , Animais , Células Cultivadas , Fibroblastos , Leucócitos Mononucleares , Camundongos
10.
Int J Dev Biol ; 63(8-9-10): 541-550, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31840791

RESUMO

Developing novel compounds for the treatment of diseases remains one of the highest priorities in biomedical research, where it is critical to identify their targets and how they work at a cellular level. Most studies in this area employ mammalian models, since rodents or non-human primates are seen as a good approximation for humans. However, using mammalian models can be problematic for a range of reasons, including high genetic redundancy and the essential role for many proteins in development. More importantly, it is very difficult to identify how compounds function at a cellular or molecular level in these models without a previously suggested mechanism or target. So how can we identify targets of medicinal compounds? In this review we outline the use of an innovative and tractable model system, Dictyostelium discoideum, to provide useful insight to the cellular and molecular functions of both therapeutic drugs and pharmacologically active natural products. We outline the advantages of using this model, and then provide a range of exemplar studies using D. discoideum in pharmacological research to demonstrate breakthroughs in understanding the action and effects of compounds, and the subsequent translational of these advances to mammalian models leading to potential improvements in societal health.


Assuntos
Produtos Biológicos/farmacologia , Dictyostelium/fisiologia , Descoberta de Drogas , Animais , Anti-Infecciosos/farmacologia , Infecções Bacterianas/tratamento farmacológico , Transtorno Bipolar/tratamento farmacológico , Cafeína/farmacologia , Dictyostelium/genética , Epilepsia/tratamento farmacológico , Flavonoides/farmacologia , Humanos , Modelos Animais , Modelos Biológicos , Neoplasias/tratamento farmacológico
11.
Dis Model Mech ; 11(1)2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29361519

RESUMO

Natural compounds often have complex molecular structures and unknown molecular targets. These characteristics make them difficult to analyse using a classical pharmacological approach. Curcumin, the main curcuminoid of turmeric, is a complex molecule possessing wide-ranging biological activities, cellular mechanisms and roles in potential therapeutic treatment, including Alzheimer's disease and cancer. Here, we investigate the physiological effects and molecular targets of curcumin in Dictyostelium discoideum We show that curcumin exerts acute effects on cell behaviour, reduces cell growth and slows multicellular development. We employed a range of structurally related compounds to show the distinct role of different structural groups in curcumin's effects on cell behaviour, growth and development, highlighting active moieties in cell function, and showing that these cellular effects are unrelated to the well-known antioxidant activity of curcumin. Molecular mechanisms underlying the effect of curcumin and one synthetic analogue (EF24) were then investigated to identify a curcumin-resistant mutant lacking the protein phosphatase 2A regulatory subunit (PsrA) and an EF24-resistant mutant lacking the presenilin 1 orthologue (PsenB). Using in silico docking analysis, we then showed that curcumin might function through direct binding to a key regulatory region of PsrA. These findings reveal novel cellular and molecular mechanisms for the function of curcumin and related compounds.


Assuntos
Curcumina/farmacologia , Dictyostelium/metabolismo , Presenilina-1/metabolismo , Proteína Fosfatase 2/metabolismo , Homologia de Sequência de Aminoácidos , Antioxidantes/farmacologia , Curcumina/análogos & derivados , Curcumina/química , Dictyostelium/efeitos dos fármacos , Dictyostelium/crescimento & desenvolvimento , Ligantes , Simulação de Acoplamento Molecular
12.
Lancet Neurol ; 17(1): 84-93, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29263011

RESUMO

High-fat, low-carbohydrate diets, known as ketogenic diets, have been used as a non-pharmacological treatment for refractory epilepsy. A key mechanism of this treatment is thought to be the generation of ketones, which provide brain cells (neurons and astrocytes) with an energy source that is more efficient than glucose, resulting in beneficial downstream metabolic changes, such as increasing adenosine levels, which might have effects on seizure control. However, some studies have challenged the central role of ketones because medium-chain fatty acids, which are part of a commonly used variation of the diet (the medium-chain triglyceride ketogenic diet), have been shown to directly inhibit AMPA receptors (glutamate receptors), and to change cell energetics through mitochondrial biogenesis. Through these mechanisms, medium-chain fatty acids rather than ketones are likely to block seizure onset and raise seizure threshold. The mechanisms underlying the ketogenic diet might also have roles in other disorders, such as preventing neurodegeneration in Alzheimer's disease, the proliferation and spread of cancer, and insulin resistance in type 2 diabetes. Analysing medium-chain fatty acids in future ketogenic diet studies will provide further insights into their importance in modified forms of the diet. Moreover, the results of these studies could facilitate the development of new pharmacological and dietary therapies for epilepsy and other disorders.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Caprilatos/metabolismo , Ácidos Decanoicos/metabolismo , Diabetes Mellitus/dietoterapia , Dieta Cetogênica/métodos , Neoplasias/dietoterapia , Convulsões/dietoterapia , Humanos
13.
Neuropharmacology ; 128: 54-62, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28947378

RESUMO

The amyloid hypothesis of Alzheimer's disease suggests that synaptic degeneration and pathology is caused by the accumulation of amyloid-ß (Aß) peptides derived from the amyloid precursor protein (APP). Subsequently, soluble Aß oligomers cause the loss of synaptic proteins from neurons, a histopathological feature of Alzheimer's disease that correlates with the degree of dementia. In this study, the production of toxic forms of Aß was examined in vitro using 7PA2 cells stably transfected with human APP. We show that conditioned media from 7PA2 cells containing Aß oligomers caused synapse degeneration as measured by the loss of synaptic proteins, including synaptophysin and cysteine-string protein, from cultured neurons. Critically, conditioned media from 7PA2 cells treated with valproic acid (2-propylpentanoic acid (VPA)) or propylisopropylacetic acid (PIA) did not cause synapse damage. Treatment with VPA or PIA did not significantly affect total Aß42 concentrations; rather these drugs selectively reduced the concentrations of Aß42 oligomers in conditioned media. In contrast, treatment significantly increased the concentrations of Aß42 monomers in conditioned media. VPA or PIA treatment reduced the concentrations of APP within lipid rafts, membrane compartments associated with Aß production. These effects of VPA and PIA were reversed by the addition of platelet-activating factor, a bioactive phospholipid produced following activation of phospholipase A2, an enzyme sensitive to VPA and PIA. Collectively these data suggest that VPA and PIA reduce Aß oligomers through inhibition of phospholipase A2 and suggest a novel therapeutic approach to Alzheimer's treatment.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Valproico/farmacologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células CHO , Estruturas da Membrana Celular/efeitos dos fármacos , Estruturas da Membrana Celular/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Colesterol/metabolismo , Cricetulus , Meios de Cultivo Condicionados/farmacologia , Embrião de Mamíferos , Epilepsia do Lobo Temporal , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fator de Ativação de Plaquetas/farmacologia , Sinaptofisina/metabolismo , Transfecção
14.
Neuropharmacology ; 101: 566-75, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26116815

RESUMO

Many neurodegenerative diseases present the loss of synapses as a common pathological feature. Here we have employed an in vitro model for synaptic loss to investigate the molecular mechanism of a therapeutic treatment, valproic acid (VPA). We show that amyloid-ß (Aß), isolated from patient tissue and thought to be the causative agent of Alzheimer's disease, caused the loss of synaptic proteins including synaptophysin, synapsin-1 and cysteine-string protein from cultured mouse neurons. Aß-induced synapse damage was reduced by pre-treatment with physiologically relevant concentrations of VPA (10 µM) and a structural variant propylisopropylacetic acid (PIA). These drugs also reduced synaptic damage induced by other neurodegenerative-associated proteins α-synuclein, linked to Lewy body dementia and Parkinson's disease, and the prion-derived peptide PrP82-146. Consistent with these effects, synaptic vesicle recycling was also inhibited by these proteins and protected by VPA and PIA. We show a mechanism for this damage through aberrant activation of cytoplasmic phospholipase A2 (cPLA2) that is reduced by both drugs. Furthermore, Aß-dependent cPLA2 activation correlates with its accumulation in lipid rafts, and is likely to be caused by elevated cholesterol (stabilising rafts) and decreased cholesterol ester levels, and this mechanism is reduced by VPA and PIA. Such observations suggest that VPA and PIA may provide protection against synaptic damage that occurs during Alzheimer's and Parkinson's and prion diseases.


Assuntos
Doença de Alzheimer/patologia , Inibidores Enzimáticos/farmacologia , Fosfolipases A2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Ácido Valproico/farmacologia , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Células Cultivadas , Colesterol/metabolismo , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Ionomicina/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Camundongos , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Príons/farmacologia , Sinapses/patologia , Sinaptofisina/metabolismo , Proteína 1 Associada à Membrana da Vesícula/metabolismo
15.
Br J Pharmacol ; 172(22): 5306-17, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26333042

RESUMO

BACKGROUND AND PURPOSE: Valproic acid (VPA), a widely used epilepsy and bipolar disorder treatment, provides acute protection against haemorrhagic shock-induced mortality in a range of in vivo models through an unknown mechanism. In the liver, this effect occurs with a concomitant protection against a decrease in GSK3ß-Ser(9) phosphorylation. Here, we developed an in vitro model to investigate this protective effect of VPA and define a molecular mechanism. EXPERIMENTAL APPROACH: The human hepatocarcinoma cell line (Huh7) was exposed to conditions occurring during haemorrhagic shock (hypoxia, hypercapnia and hypothermia) to investigate the changes in GSK3ß-Ser(9) phosphorylation for a 4 h period following treatment with VPA, related congeners, PPAR agonists, antagonists and siRNA. KEY RESULTS: Huh7 cells undergoing combined hypoxia, hypercapnia, and hypothermia reproduced the reduced GSK3ß-Ser(9) phosphorylation shown in vivo during haemorrhagic shock, and this change was blocked by VPA. The protective effect occurred through upstream PTEN and Akt signalling, and prevented downstream ß-catenin degradation while increasing histone 2/3 acetylation. This effect was reproduced by several VPA-related compounds with known PPARγ agonist activity, independent of histone deacetylase (HDAC) inhibitory activity. Specific pharmacological inhibition (by T0070907) or knockdown of PPARγ blocked the protective effect of VPA against these signalling changes and apoptosis. In addition, specific activation of PPARγ using ciglitazone reproduced the changes induced by VPA in haemorrhagic shock-like conditions. CONCLUSION AND IMPLICATIONS: Changes in GSK3ß-Ser(9) phosphorylation in in vivo haemorrhagic shock models can be modelled in vitro, and this has identified a role for PPARγ activation in the protective role of VPA.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , PPAR gama/metabolismo , Substâncias Protetoras/farmacologia , Choque Hemorrágico/metabolismo , Ácido Valproico/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Glicogênio Sintase Quinase 3 beta , Humanos , Hipercapnia/metabolismo , Hipotermia/metabolismo , Hipóxia/metabolismo , L-Lactato Desidrogenase/metabolismo , PPAR gama/agonistas , PPAR gama/genética , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , Transdução de Sinais
16.
Neurobiol Dis ; 62: 296-306, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24148856

RESUMO

Phosphatidylinositol (3-5) trisphosphate (PIP3) is a central regulator of diverse neuronal functions that are critical for seizure progression, however its role in seizures is unclear. We have recently hypothesised that valproic acid (VPA), one of the most commonly used drugs for the treatment of epilepsy, may target PIP3 signalling as a therapeutic mode of action. Here, we show that seizure induction using kainic acid in a rat in vivo epilepsy model resulted in a decrease in hippocampal PIP3 levels and reduced protein kinase B (PKB/AKT) phosphorylation, measured using ELISA mass assays and Western blot analysis, and both changes were restored following VPA treatment. These finding were reproduced in cultured rat hippocampal primary neurons and entorhinal cortex-hippocampal slices during exposure to the GABA(A) receptor antagonist pentylenetetrazol (PTZ), which is widely used to generate seizures and seizure-like (paroxysmal) activity. Moreover, VPA's effect on paroxysmal activity in the PTZ slice model is blocked by phosphatidylinositol 3-kinase (PI3K) inhibition or PIP2 sequestration by neomycin, indicating that VPA's efficacy is dependent upon PIP3 signalling. PIP3 depletion following PTZ treatment may also provide a positive feedback loop, since enhancing PIP3 depletion increases, and conversely, reducing PIP3 dephosphorylation reduces paroxysmal activity and this effect is dependent upon AMPA receptor activation. Our results therefore indicate that PIP3 depletion occurs with seizure activity, and that VPA functions to reverse these effects, providing a novel mechanism for VPA in epilepsy treatment.


Assuntos
Anticonvulsivantes/uso terapêutico , Neurônios/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/metabolismo , Ácido Valproico/uso terapêutico , Animais , Células Cultivadas , Antagonistas GABAérgicos/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Ácido Caínico/toxicidade , Masculino , Pentilenotetrazol/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Estado Epiléptico/induzido quimicamente
17.
J Cell Sci ; 126(Pt 23): 5465-76, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24006265

RESUMO

Detection of substances tasting bitter to humans occurs in diverse organisms including the social amoeba Dictyostelium discoideum. To establish a molecular mechanism for bitter tastant detection in Dictyostelium, we screened a mutant library for resistance to a commonly used bitter standard, phenylthiourea. This approach identified a G-protein-coupled receptor mutant, grlJ(-), which showed a significantly increased tolerance to phenylthiourea in growth, survival and movement. This mutant was not resistant to a structurally dissimilar potent bitter tastant, denatonium benzoate, suggesting it is not a target for at least one other bitter tastant. Analysis of the cell-signalling pathway involved in the detection of phenylthiourea showed dependence upon heterotrimeric G protein and phosphatidylinositol 3-kinase activity, suggesting that this signalling pathway is responsible for the cellular effects of phenylthiourea. This is further supported by a phenylthiourea-dependent block in the transient cAMP-induced production of phosphatidylinositol (3,4,5)-trisphosphate (PIP3) in wild-type but not grlJ(-) cells. Finally, we have identified an uncharacterized human protein γ-aminobutyric acid (GABA) type B receptor subunit 1 isoform with weak homology to GrlJ that restored grlJ(-) sensitivity to phenylthiourea in cell movement and PIP3 regulation. Our results thus identify a novel pathway for the detection of the standard bitter tastant phenylthiourea in Dictyostelium and implicate a poorly characterized human protein in phenylthiourea-dependent cell responses.


Assuntos
Dictyostelium/fisiologia , Feniltioureia/química , Fosfatidilinositol 3-Quinase/genética , Receptores Acoplados a Proteínas G/genética , Receptores de GABA-B/genética , Paladar/fisiologia , Movimento Celular , Sobrevivência Celular , AMP Cíclico/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Teste de Complementação Genética , Humanos , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Compostos de Amônio Quaternário/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores de GABA-B/metabolismo , Transdução de Sinais , Papilas Gustativas/metabolismo
18.
PLoS One ; 5(6): e11151, 2010 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-20567601

RESUMO

Lithium (Li(+)) is a common treatment for bipolar mood disorder, a major psychiatric illness with a lifetime prevalence of more than 1%. Risk of bipolar disorder is heavily influenced by genetic predisposition, but is a complex genetic trait and, to date, genetic studies have provided little insight into its molecular origins. An alternative approach is to investigate the genetics of Li(+) sensitivity. Using the social amoeba Dictyostelium, we previously identified prolyl oligopeptidase (PO) as a modulator of Li(+) sensitivity. In a link to the clinic, PO enzyme activity is altered in bipolar disorder patients. Further studies demonstrated that PO is a negative regulator of inositol(1,4,5)trisphosphate (IP(3)) synthesis, a Li(+) sensitive intracellular signal. However, it was unclear how PO could influence either Li(+) sensitivity or risk of bipolar disorder. Here we show that in both Dictyostelium and cultured human cells PO acts via Multiple Inositol Polyphosphate Phosphatase (Mipp1) to control gene expression. This reveals a novel, gene regulatory network that modulates inositol metabolism and Li(+) sensitivity. Among its targets is the inositol monophosphatase gene IMPA2, which has also been associated with risk of bipolar disorder in some family studies, and our observations offer a cellular signalling pathway in which PO activity and IMPA2 gene expression converge.


Assuntos
Resistência a Medicamentos/genética , Regulação da Expressão Gênica , Inositol/biossíntese , Compostos de Lítio/farmacologia , Quimiotaxia/efeitos dos fármacos , Dictyostelium/genética , Monoéster Fosfórico Hidrolases/metabolismo
19.
Epilepsia ; 51(8): 1533-42, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20002144

RESUMO

PURPOSE: Valproic acid (VPA) is widely used clinically in epilepsy, bipolar disorder, and migraine. In experimental models, it has also been shown to have neuroprotective and antiepileptogenic effects. Its mechanisms of action in these diverse conditions are, however, unclear, but there is some evidence indicating an effect of VPA upon protein kinase A (PKA) activity. We, therefore, asked whether VPA modulates cyclic adenosine monophosphate (cAMP)/PKA-dependent synaptic plasticity and whether this mode of action could explain its anticonvulsant effect. METHODS: We first tested the effects of VPA on PKA-dependent synaptic plasticity at mossy fiber to CA3 synapses in rat hippocampus slices following very high-frequency stimulation or application of the adenylyl cyclase activator forskolin. Using biochemical assays, we then tested whether VPA had a direct effect on PKA activity or an indirect effect through modulating cAMP production. Lastly, VPA and inhibitors of adenylyl cyclase (SQ22536) and PKA (H89) were tested in in vitro models of epileptiform activity induced in hippocampal-entorhinal cortex slices using either pentylenetetrazol (2 mM) or low magnesium. RESULTS: VPA (1 mm) inhibited PKA-dependent long-term potentiation of mossy fiber to CA3 pyramidal cell transmission. However, VPA did not directly modulate PKA activity but rather inhibited the accumulation of cAMP. In acute in vitro seizure models, the anticonvulsant activity of VPA is not mediated through modulation of adenylyl cyclase or PKA. CONCLUSIONS: These results indicate that VPA through an action on cAMP accumulation can inhibit synaptic plasticity, but this cannot fully explain its anticonvulsant effect.


Assuntos
Anticonvulsivantes/farmacologia , Encéfalo , AMP Cíclico/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Ácido Valproico/farmacologia , Análise de Variância , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Epilepsia/patologia , Isoquinolinas/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia
20.
Biochem Soc Trans ; 37(Pt 5): 1110-4, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19754462

RESUMO

Lithium (Li(+)) is the mood stabilizer most frequently used in the treatment of bipolar mood disorder; however, its therapeutic mechanism is unknown. In the 1980s, Berridge and colleagues proposed that Li(+) treatment acts via inhibition of IMPase (inositol monophosphatase) to deplete the cellular concentration of myo-inositol. Inositol depletion is also seen with the alternative mood stabilizers VPA (valproic acid) and CBZ (carbamazepine), suggesting a common therapeutic action. All three drugs cause changes in neuronal cell morphology and cell chemotaxis; however, it is unclear how reduced cellular inositol modulates these changes in cell behaviour. It is often assumed that reduced inositol suppresses Ins(1,4,5)P(3), a major intracellular signal molecule, but there are other important phosphoinostide-based signal molecules in the cell. In the present paper, we discuss evidence that Li(+) has a substantial effect on PtdIns(3,4,5)P(3), an important signal molecule within the nervous system. As seen for Ins(1,4,5)P(3) signalling, suppression of PtdIns(3,4,5)P(3) signalling also occurs via an inositol-depletion mechanism. This has implications for the cellular mechanisms controlling phosphoinositide signalling, and offers insight into the genetics underlying risk of bipolar mood disorder.


Assuntos
Transtorno Bipolar/tratamento farmacológico , Inositol/metabolismo , Compostos de Lítio , Fosfatos de Fosfatidilinositol/metabolismo , Animais , Antimaníacos/farmacologia , Antimaníacos/uso terapêutico , Transtorno Bipolar/genética , Transtorno Bipolar/fisiopatologia , Carbamazepina/farmacologia , Carbamazepina/uso terapêutico , Quimiotaxia/efeitos dos fármacos , Humanos , Compostos de Lítio/farmacologia , Compostos de Lítio/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ácido Valproico/farmacologia , Ácido Valproico/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA