Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 34(7): 1522-30, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24855057

RESUMO

OBJECTIVE: Voltage-dependent K(+) (Kv) channels from the Kv7 family are expressed in blood vessels and contribute to cardiovascular physiology. Although Kv7 channel blockers trigger muscle contractions, Kv7 activators act as vasorelaxants. Kv7.1 and Kv7.5 are expressed in many vessels. Kv7.1 is under intense investigation because Kv7.1 blockers fail to modulate smooth muscle reactivity. In this study, we analyzed whether Kv7.1 and Kv7.5 may form functional heterotetrameric channels increasing the channel diversity in vascular smooth muscles. APPROACH AND RESULTS: Kv7.1 and Kv7.5 currents elicited in arterial myocytes, oocyte, and mammalian expression systems suggest the formation of heterotetrameric complexes. Kv7.1/Kv7.5 heteromers, exhibiting different pharmacological characteristics, participate in the arterial tone. Kv7.1/Kv7.5 associations were confirmed by coimmunoprecipitation, fluorescence resonance energy transfer, and fluorescence recovery after photobleaching experiments. Kv7.1/Kv7.5 heterotetramers were highly retained at the endoplasmic reticulum. Studies in HEK-293 cells, heart, brain, and smooth and skeletal muscles demonstrated that the predominant presence of Kv7.5 stimulates release of Kv7.1/Kv7.5 oligomers out of lipid raft microdomains. Electrophysiological studies supported that KCNE1 and KCNE3 regulatory subunits further increased the channel diversity. Finally, the analysis of rat isolated myocytes and human blood vessels demonstrated that Kv7.1 and Kv7.5 exhibited a differential expression, which may lead to channel diversity. CONCLUSIONS: Kv7.1 and Kv7.5 form heterotetrameric channels increasing the diversity of structures which fine-tune blood vessel reactivity. Because the lipid raft localization of ion channels is crucial for cardiovascular physiology, Kv7.1/Kv7.5 heteromers provide efficient spatial and temporal regulation of smooth muscle function. Our results shed light on the debate about the contribution of Kv7 channels to vasoconstriction and hypertension.


Assuntos
Canais de Potássio KCNQ/metabolismo , Canal de Potássio KCNQ1/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Potássio/metabolismo , Animais , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Canais de Potássio KCNQ/química , Canais de Potássio KCNQ/efeitos dos fármacos , Canais de Potássio KCNQ/genética , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ1/genética , Microdomínios da Membrana/metabolismo , Potenciais da Membrana , Músculo Liso Vascular/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Estrutura Quaternária de Proteína , Ratos , Transfecção , Xenopus
2.
Muscle Nerve ; 45(1): 48-54, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22190306

RESUMO

BACKGROUND: Kv7.5 (KCNQ5) channels conduct M-type potassium currents in the brain, are expressed in skeletal muscle, and contribute to vascular muscle tone. METHODS: We coexpressed Kv7.5 and KCNE1-3 peptides in HEK293 cells and then analyzed their association using electrophysiology and co-immunoprecipitation, assessed localization using confocal microscopy, examined targeting of the oligomeric channels to cholesterol-rich membrane surface microdomains using lipid raft isolation, and evaluated their membrane dynamics using fluorescence recovery after photobleaching (FRAP). RESULTS: Kv7.5 forms oligomeric channels specifically with KCNE1 and KCNE3. The expression of Kv7.5 targeted to cholesterol-rich membrane surface microdomains was very low. Oligomeric Kv7.5/KCNE1 and Kv7.5/KCNE3 channels did not localize to lipid rafts. However, Kv7.5 association impaired KCNE3 expression in lipid raft microdomains. CONCLUSIONS: Our results indicate that Kv7.5 contributes to the spatial regulation of KCNE3. This new scenario could greatly assist in determining the physiological relevance of putative KCNE3 interactions in nerve and muscle.


Assuntos
Canais de Potássio KCNQ/metabolismo , Microdomínios da Membrana/metabolismo , Potenciais da Membrana/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Linhagem Celular Transformada , Estimulação Elétrica , Recuperação de Fluorescência Após Fotodegradação , Humanos , Imunoprecipitação , Canais de Potássio KCNQ/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transfecção/métodos
3.
J Neurosci ; 30(27): 9316-23, 2010 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-20610766

RESUMO

KCNQ2 (Kv7.2) and KCNQ3 (Kv7.3) are the principal subunits underlying the potassium M-current, which exerts a strong control on neuronal excitability. KCNQ3 subunits coassemble with KCNQ2 to form functional heteromeric channels that are specifically transported to the axonal initial segment and nodes of Ranvier. In contrast, there is no evidence for functional homomeric KCNQ3 channels in neurons, and it appears that these are inefficiently trafficked to the plasma membrane. Among eukaryotic potassium channels, the KCNQ3 subunit is unusual because it has an alanine in place of a threonine at the pore inner vestibule, three residues upstream of the GYG signature sequence of the selectivity filter. This residue is critical for the potentiation of the current after heteromerization, but the mechanism is unknown. We report that the presence of this uncommon residue at position 315 has a strong impact on the stability of the homotetramers and on channel trafficking. Wild-type KCNQ3 expressed alone is retained within the endoplasmic reticulum, and this mechanism is overcome by the substitution of threonine for Ala315. KCNQ3 subunits require assembly with KCNQ2 to exit this compartment, whereas KCNQ3-A315T is no longer dependent on KCNQ2 to form channels that are efficiently trafficked to the plasma membrane. The presence of this alanine, therefore, plays an important role in regulating the subunit composition of functional M-channels expressed at the surface of neurons.


Assuntos
Membrana Celular/metabolismo , Regulação da Expressão Gênica/fisiologia , Canal de Potássio KCNQ3/química , Canal de Potássio KCNQ3/metabolismo , Alanina/metabolismo , Substituição de Aminoácidos/genética , Animais , Proteínas de Bactérias/genética , Linhagem Celular Transformada , Membrana Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Humanos , Imunoprecipitação/métodos , Ativação do Canal Iônico/genética , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/genética , Proteínas Luminescentes/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Oócitos , Técnicas de Patch-Clamp/métodos , Bloqueadores dos Canais de Potássio/farmacologia , Relação Estrutura-Atividade , Tetraetilamônio/farmacocinética , Transdução Genética/métodos , Xenopus
4.
J Cell Sci ; 122(Pt 20): 3738-48, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19773357

RESUMO

Voltage-dependent potassium channels (Kv) play a crucial role in the activation and proliferation of leukocytes. Kv channels are either homo- or hetero-oligomers. This composition modulates their surface expression and serves as a mechanism for regulating channel activity. Kv channel interaction with accessory subunits provides mechanisms for channels to respond to stimuli beyond changes in membrane potential. Here, we demonstrate that KCNE4 (potassium voltage-gated channel subfamily E member 4), but not KCNE2, functions as an inhibitory Kv1.3 partner in leukocytes. Kv1.3 trafficking, targeting and activity are altered by the presence of KCNE4. KCNE4 decreases current density, slows activation, accelerates inactivation, increases cumulative inactivation, retains Kv1.3 in the ER and impairs channel targeting to lipid raft microdomains. KCNE4 associates with Kv1.3 in the ER and decreases the number of Kv1.3 channels at the cell surface, which diminishes cell excitability. Kv1.3 and KCNE4 are differentially regulated upon activation or immunosuppression in macrophages. Thus, lipopolysaccharide-induced activation increases Kv1.3 and KCNE4 mRNA, whereas dexamethasone triggers a decrease in Kv1.3 with no changes in KCNE4. The channelosome composition determines the activity and affects surface expression and membrane localization. Therefore, KCNE4 association might play a crucial role in controlling immunological responses. Our results indicate that KCNE ancillary subunits could be new targets for immunomodulation.


Assuntos
Membrana Celular/metabolismo , Ativação do Canal Iônico , Canal de Potássio Kv1.3/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Regulação da Expressão Gênica , Humanos , Macrófagos/metabolismo , Camundongos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Ligação Proteica , Estrutura Quaternária de Proteína , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos
5.
J Cell Physiol ; 225(3): 692-700, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20533308

RESUMO

The KCNQ1 (Kv7.1) channel plays an important role in cardiovascular physiology. Cardiomyocytes co-express KCNQ1 with KCNE1-5 proteins. KCNQ1 may co-associate with multiple KCNE regulatory subunits to generate different biophysically and pharmacologically distinct channels. Increasing evidence indicates that the location and targeting of channels are important determinants of their function. In this context, the presence of K(+) channels in sphingolipid-cholesterol-enriched membrane microdomains (lipid rafts) is under investigation. Lipid rafts are important for cardiovascular functioning. We aimed to determine whether KCNE subunits modify the localization and targeting of KCNQ1 channels in lipid rafts microdomains. HEK-293 cells were transiently transfected with KCNQ1 and KCNE1-5, and their traffic and presence in lipid rafts were analyzed. Only KCNQ1 and KCNE3, when expressed alone, co-localized in raft fractions. In addition, while KCNE2 and KCNE5 notably stained the cell surface, KCNQ1 and the rest of the KCNEs showed strong intracellular retention. KCNQ1 targets multiple membrane surface microdomains upon association with KCNE peptides. Thus, while KCNQ1/KCNE1 and KCNQ1/KCNE2 channels target lipid rafts, KCNQ1 associated with KCNE3-5 did not. Channel membrane dynamics, analyzed by fluorescence recovery after photobleaching (FRAP) experiments, further supported these results. In conclusion, the trafficking and targeting pattern of KCNQ1 can be influenced by its association with KCNEs. Since KCNQ1 is crucial for cardiovascular physiology, the temporal and spatial regulations that different KCNE subunits may confer to the channels could have a dramatic impact on membrane electrical activity and putative endocrine regulation.


Assuntos
Canal de Potássio KCNQ1/metabolismo , Microdomínios da Membrana/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Linhagem Celular , Recuperação de Fluorescência Após Fotodegradação , Humanos , Canal de Potássio KCNQ1/genética , Cinética , Microscopia Confocal , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Subunidades Proteicas , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
6.
Biochim Biophys Acta ; 1783(5): 728-36, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18230363

RESUMO

Voltage-dependent K(+) channels (Kv) are involved in the proliferation of many types of cells, but the mechanisms by which their activity is related to cell growth remain unclear. Kv antagonists inhibit the proliferation of mammalian cells, which is of physiological relevance in skeletal muscle. Although myofibres are terminally differentiated, some resident myoblasts may re-enter the cell cycle and proliferate. Here we report that the expression of Kv1.5 is cell-cycle dependent during myoblast proliferation. In addition to Kv1.5 other Kv, such as Kv1.3, are also up-regulated. However, pharmacological evidence mainly implicates Kv1.5 in myoblast growth. Thus, the presence of S0100176, a Kv antagonist, but not margatoxin and dendrotoxin, led to cell cycle arrest during the G(1)-phase. The use of selective cell cycle blockers showed that Kv1.5 was transiently accumulated during the early G(1)-phase. Furthermore, while myoblasts treated with S0100176 expressed low levels of cyclin A and D(1), the expression of p21(cip-1) and p27(kip1), two cyclin-dependent kinase inhibitors, increased. Our results indicate that the cell cycle-dependent expression of Kv1.5 is involved in skeletal muscle cell proliferation.


Assuntos
Ciclo Celular , Canal de Potássio Kv1.5/metabolismo , Mioblastos Esqueléticos/metabolismo , Animais , Ciclo Celular/genética , Linhagem Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Expressão Gênica , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/fisiologia , Mioblastos Esqueléticos/citologia , Ratos
7.
Cell Physiol Biochem ; 24(5-6): 325-34, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19910673

RESUMO

Kv7 (KCNQ) proteins form a family of voltage-gated potassium channels that is comprised of five members, Kv7.1-Kv7.5. While Kv7.1 is crucial in the heart, the Kv7.2, Kv7.3, Kv7.4 and Kv7.5 channels contribute to the M-current in the nervous system. In addition to the brain, Kv7.5 is expressed in skeletal and smooth muscle, where its physiological role is currently under evaluation. Kv7 associations with KCNE accessory subunits (KCNE1-5) enhance channel diversity and their interaction provides mechanisms to respond to a variety of stimuli. KCNE peptides control the surface expression, voltage-dependence, kinetics of gating, unitary conductance, ion selectivity and pharmacology of several channels. KCNE subunits have been primarily studied in the heart; however, their activity in the brain and in many other tissues is being increasingly recognized. Here, we found that Kv7.5 and KCNE subunits are present in myoblasts. Therefore, oligomeric associations may underlie some Kv7.5 functional diversity in skeletal muscle. An extensive study in Xenopus oocytes and HEK-293 cells demonstrates that KCNE1 and KCNE3, but none of the other KCNE subunits, affect Kv7.5 currents. While KCNE1 slows activation and suppresses inward rectification, KCNE3 drastically inhibits Kv7.5 currents. In addition, KCNE1 increases Kv7.5 currents in HEK cells. Changes in gating and amplitude indicate functional interactions. Our results have physiological relevance since Kv7.5 is abundant in skeletal and smooth muscle and its association with KCNE peptides may fine-tune cellular responses.


Assuntos
Canais de Potássio KCNQ/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Linhagem Celular , Fenômenos Eletrofisiológicos , Humanos , Canais de Potássio KCNQ/genética , Músculo Esquelético/metabolismo , Oócitos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Fase S , Xenopus
8.
Sci Rep ; 9(1): 11920, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31417177

RESUMO

Extracellular vesicles (EVs) are important mediators of cell-cell communication in a broad variety of physiological contexts. However, there is ambiguity around the fundamental mechanisms by which these effects are transduced, particularly in relation to their uptake by recipient cells. Multiple modes of cellular entry have been suggested and we have further explored the role of glycans as potential determinants of uptake, using EVs from the murine hepatic cell lines AML12 and MLP29 as independent yet comparable models. Lectin microarray technology was employed to define the surface glycosylation patterns of EVs. Glycosidases PNGase F and neuraminidase which cleave N-glycans and terminal sialic acids, respectively, were used to analyze the relevance of these modifications to EV surface glycans on the uptake of fluorescently labelled EVs by a panel of cells representing a variety of tissues. Flow cytometry revealed an increase in affinity for EVs modified by both glycosidase treatments. High-content screening exhibited a broader range of responses with different cell types preferring different vesicle glycosylation states. We also found differences in vesicle charge after treatment with glycosidases. We conclude that glycans are key players in the tuning of EV uptake, through charge-based effects, direct glycan recognition or both, supporting glycoengineering as a toolkit for therapy development.


Assuntos
Endocitose , Vesículas Extracelulares/metabolismo , Polissacarídeos/metabolismo , Linhagem Celular , Vesículas Extracelulares/ultraestrutura , Glicosídeo Hidrolases/metabolismo , Glicosilação , Humanos , Lectinas/metabolismo , Neuraminidase/metabolismo , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/metabolismo
9.
Biochem Biophys Res Commun ; 369(4): 1094-7, 2008 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-18331828

RESUMO

Voltage-dependent K(+) channels (Kv) are involved in myocyte proliferation and differentiation by triggering changes in membrane potential and regulating cell volume. Since Kv7 channels may participate in these events, the purpose of this study was to investigate whether skeletal muscle Kv7.1 and Kv7.5 were involved during proliferation and myogenesis. Here we report that, while myotube formation did not regulate Kv7 channels, Kv7.5 was up-regulated during cell cycle progression. Although, Kv7.1 mRNA also increased during the G(1)-phase, pharmacological evidence mainly involves Kv7.5 in myoblast growth. Our results indicate that the cell cycle-dependent expression of Kv7.5 is involved in skeletal muscle cell proliferation.


Assuntos
Ciclo Celular , Diferenciação Celular , Canais de Potássio KCNQ/metabolismo , Canal de Potássio KCNQ1/metabolismo , Desenvolvimento Muscular , Mioblastos Esqueléticos/citologia , Animais , Linhagem Celular , Proliferação de Células , Canais de Potássio KCNQ/genética , Canal de Potássio KCNQ1/genética , Desenvolvimento Muscular/genética , Mioblastos Esqueléticos/metabolismo , Ratos
10.
SLAS Technol ; 23(3): 207-216, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29412765

RESUMO

Nomad Technology (Innoprot [Innovative Technologies in Biological Systems], Derio, Spain), a novel tool for multiplexing high-throughput cell-based G protein-coupled receptor (GPCR) assays, is described in this work. This new technology comprises a family of fluorescent biosensors called Nomad Biosensors that allow for the measurement of responses mediated by G proteins through their interactions with second-messenger transduction proteins. GPCRs are one of the largest protein families of receptors in eukaryotes, and their signaling mediates important physiological processes within cells. Thus, GPCRs are associated with a wide variety of diseases, and considered major targets in therapeutic research. Nomad constitutes a novel tool for unraveling the mechanism of GPCR signal transduction by simultaneously tracing different pathways. GPCR activation changes the structural folding of the biosensor and promotes its vesicularization, as well as an increase in the fluorescence intensity. Based on this technology, the MPXNomad cellular model was developed to discriminate between the Ca2+-mediated pathway and the cyclic adenosine monophosphate (cAMP)-mediated pathway. To validate this model, endothelin receptor B (ETBR) was coexpressed into the MPXNomad cell line and assessed with a specific agonist, an antagonist, and a chemical library of compounds. Nomad Technology optimizes the identification of novel GPCR ligands and enables the testing of large numbers of compounds.


Assuntos
Técnicas Biossensoriais , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Cálcio/metabolismo , Linhagem Celular Tumoral , Clonagem Molecular , AMP Cíclico/metabolismo , Endotelinas/metabolismo , Fluorescência , Humanos , Processamento de Imagem Assistida por Computador , Ligantes , Receptor de Endotelina B/genética , Receptor de Endotelina B/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais
11.
PLoS Negl Trop Dis ; 12(1): e0006180, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357372

RESUMO

Chagas disease remains one of the most neglected diseases in the world despite being the most important parasitic disease in Latin America. The characteristic chronic manifestation of chagasic cardiomyopathy is the region's leading cause of heart-related illness, causing significant mortality and morbidity. Due to the limited available therapeutic options, new drugs are urgently needed to control the disease. Sirtuins, also called Silent information regulator 2 (Sir2) proteins have long been suggested as interesting targets to treat different diseases, including parasitic infections. Recent studies on Trypanosoma cruzi sirtuins have hinted at the possibility to exploit these enzymes as a possible drug targets. In the present work, the T. cruzi Sir2 related protein 1 (TcSir2rp1) is genetically validated as a drug target and biochemically characterized for its NAD+-dependent deacetylase activity and its inhibition by the classic sirtuin inhibitor nicotinamide, as well as by bisnaphthalimidopropyl (BNIP) derivatives, a class of parasite sirtuin inhibitors. BNIPs ability to inhibit TcSir2rp1, and anti-parasitic activity against T. cruzi amastigotes in vitro were investigated. The compound BNIP Spermidine (BNIPSpd) (9), was found to be the most potent inhibitor of TcSir2rp1. Moreover, this compound showed altered trypanocidal activity against TcSir2rp1 overexpressing epimastigotes and anti-parasitic activity similar to the reference drug benznidazole against the medically important amastigotes, while having the highest selectivity index amongst the compounds tested. Unfortunately, BNIPSpd failed to treat a mouse model of Chagas disease, possibly due to its pharmacokinetic profile. Medicinal chemistry modifications of the compound, as well as alternative formulations may improve activity and pharmacokinetics in the future. Additionally, an initial TcSIR2rp1 model in complex with p53 peptide substrate was obtained from low resolution X-ray data (3.5 Å) to gain insight into the potential specificity of the interaction with the BNIP compounds. In conclusion, the search for TcSir2rp1 specific inhibitors may represent a valuable strategy for drug discovery against T. cruzi.


Assuntos
Antiprotozoários/metabolismo , Doença de Chagas/parasitologia , Inibidores Enzimáticos/metabolismo , Proteínas de Protozoários/antagonistas & inibidores , Trypanosoma cruzi/efeitos dos fármacos , Animais , Doença de Chagas/tratamento farmacológico , Modelos Animais de Doenças , Camundongos , Niacinamida/metabolismo , Quinolonas/metabolismo , Espermina/análogos & derivados , Espermina/metabolismo , Resultado do Tratamento
12.
SLAS Discov ; 22(1): 67-76, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27703082

RESUMO

Parkinson disease (PD) is a prevalent neurodegenerative disease characterized by selective degeneration of dopaminergic neurons in the substantia nigra, causing tremor and motor impairment. Parkin protein, whose mutants are the cause of Parkinson disease type 2 (PARK2), has been mechanistically linked to the regulation of apoptosis and the turnover of damaged mitochondria. Several studies have implicated aberrant mitochondria as a key contributor to the development of PD. In the attempt to discover new drugs, high-content cell-based assays are becoming more important to mimic the nature of biological processes and their diversifications in diseases and will be essential for lead identification and the optimization of therapeutic candidates. We have developed a novel fluorescence cell-based assay for high-content screening to find compounds that can promote the mitochondrial localization of Parkin without severe mitochondrial damage induction. In this work, this model was used to screen a library of 1280 compounds. After the screening campaign, the positive compounds were chosen for further testing, based on the strength of the initial response and lack of cytotoxicity. These results indicated that this Parkin cell-based assay is a robust (Z' > 0.5) and valid strategy to test potential candidates for preclinical studies.


Assuntos
Bioensaio/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Doença de Parkinson/tratamento farmacológico , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Tumoral , Fluorescência , Humanos , Mitocôndrias/metabolismo , Doença de Parkinson/metabolismo , Proscilaridina/uso terapêutico , Rodaminas
13.
J Inorg Biochem ; 149: 12-24, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25957189

RESUMO

Naringenin, a natural antioxidant present in grapefruit, oranges and the skin of tomatoes showed low antioxidant properties among other flavonoids due to its structural characteristics. Since many flavonoids were shown to have cell-killing and antioxidant activities, naringenin was investigated herein. In parallel with its antioxidant activities the flavonoid showed very low cytotoxicity at concentrations up to 100 µM against lung (A549) and breast (SKBr3 and MDAMB231) cancer cell lines. Furthermore, a newly-synthesized and characterized complex of naringenin and oxidovanadium(IV) ([V(IV)O(nar)2] · 2H2O, VOnar, with weak ferromagnetic coupling) was also studied. As a result, VOnar acted as a better compound on cell-killing and antioxidant activities (in vitro) than naringenin. The anti-proliferative effect of VOnar was accompanied by reactive oxygen species (ROS) generation, cell membrane and DNA damages, cell cycle arrest, caspase 3/7 activation and mitochondrial potential reduction. The higher parameters observed for the MDAMB231 cell line have been related to its low glutathione (GSH) content. The assays of the interaction of bovine serum albumin (BSA) with the complex showed the affinity of protein toward it and that there is only one binding site on the BSA molecule. However, metal complexation decreased the binding affinity to BSA of naringenin probably due to a steric hindrance of the complex.


Assuntos
Antineoplásicos/farmacologia , Antioxidantes/farmacologia , Flavanonas/química , Compostos Organometálicos/farmacologia , Soroalbumina Bovina/metabolismo , Vanádio/química , Antineoplásicos/química , Antioxidantes/química , Apoptose , Sítios de Ligação , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Dano ao DNA , Células Epiteliais/efeitos dos fármacos , Humanos , Compostos Organometálicos/química , Ligação Proteica , Soroalbumina Bovina/química
14.
J Inorg Biochem ; 135: 86-99, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24681549

RESUMO

A new chlorogenate oxidovanadium complex (Na[VO(chlorog)(H2O)3].4H2O) was synthesized by using Schlenk methodology in the course of a reaction at inert atmosphere in which deprotonated chlorogenic acid ligand binds to oxidovanadium(IV) in a reaction experiment controlled via EPR technique and based in a species distribution diagram. The compound was characterized by FTIR, EPR, UV-visible and diffuse reflectance spectroscopies and thermogravimetric, differential thermal and elemental analyses. The ligand and the complex were tested for their antioxidant effects on DPPH (1,1-diphenyl-2-picrylhydrazyl radical), ABTS(+) (radical cation of 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt), O2(-), OH and ROO radicals and their cytotoxic activity on different cancer cell lines (SKBR3, T47D and MDAMB231) and primary human mammary epithelial cells. The complex behaved as good antioxidant agent with strongest inhibitory effects on O2(-), OH and ROO radicals and exhibited selective cytotoxicity against SKBR3 cancer cell line. Albumin interaction experiments denoted high affinity toward the complex and its calculated binding constant was indicative of a strong binding to the protein. Based on this study, it is hypothesized that Na[VO(chlorog)(H2O)3].4H2O would be a promising candidate for further evaluation as an antioxidant and anticancer agent.


Assuntos
Antineoplásicos/síntese química , Complexos de Coordenação/síntese química , Sequestradores de Radicais Livres/síntese química , Soroalbumina Bovina/química , Animais , Antineoplásicos/farmacologia , Compostos de Bifenilo/química , Neoplasias da Mama , Bovinos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Complexos de Coordenação/farmacologia , Estabilidade de Medicamentos , Feminino , Sequestradores de Radicais Livres/farmacologia , Radicais Livres/química , Humanos , Oxirredução , Picratos/química , Ligação Proteica , Superóxido Dismutase/química
15.
PLoS One ; 6(9): e25508, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21980481

RESUMO

M-channels are voltage-gated potassium channels composed of Kv7.2-7.5 subunits that serve as important regulators of neuronal excitability. Calmodulin binding is required for Kv7 channel function and mutations in Kv7.2 that disrupt calmodulin binding cause Benign Familial Neonatal Convulsions (BFNC), a dominantly inherited human epilepsy. On the basis that Kv7.2 mutants deficient in calmodulin binding are not functional, calmodulin has been defined as an auxiliary subunit of Kv7 channels. However, we have identified a presumably phosphomimetic mutation S511D that permits calmodulin-independent function. Thus, our data reveal that constitutive tethering of calmodulin is not required for Kv7 channel function.


Assuntos
Calmodulina/metabolismo , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Canal de Potássio KCNQ2/química , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/química , Canal de Potássio KCNQ3/genética , Dados de Sequência Molecular , Mutação , Estrutura Secundária de Proteína , Transporte Proteico
16.
Recent Pat Cardiovasc Drug Discov ; 3(2): 105-18, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18537761

RESUMO

Potassium channels, which are essential to a wide range of physiological processes, are involved in many diseases. Thus, alterations in such important proteins due to congenital deficiencies or to undesirable side-effects of common medications might lead to dysfunctions. Heart is one of those tissues where potassium channels play a crucial role. The maintenance of cardiac action potential appears to be the consequence of the varied activity of several types of potassium channels. Recently, compounds that modify cardiac potassium channel activity and so alter action potential duration have been developed as new anti-arrhythmic agents. However, several cardiomyopathies appear as undesirable side-effects of the use of drugs that directly or indirectly act on the same potassium channels. Thus, new patents have been created allowing the prediction of the inherited predisposition to any known potassium-linked cardiac channelopathy.


Assuntos
Antiarrítmicos/efeitos adversos , Arritmias Cardíacas/induzido quimicamente , Cardiomiopatias/induzido quimicamente , Sistema de Condução Cardíaco/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/efeitos adversos , Canais de Potássio/efeitos dos fármacos , Potenciais de Ação , Animais , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Predisposição Genética para Doença , Testes Genéticos , Sistema de Condução Cardíaco/metabolismo , Humanos , Patentes como Assunto , Canais de Potássio/genética , Canais de Potássio/metabolismo , Medição de Risco , Fatores de Risco
17.
Cardiovasc Res ; 76(3): 430-41, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17900547

RESUMO

OBJECTIVE: Cellular cardiomyoplasty using skeletal myoblasts is a promising therapy for myocardial infarct repair. Once transplanted, myoblasts grow, differentiate and adapt their electrophysiological properties towards more cardiac-like phenotypes. Voltage-dependent Na(+) channels (Na(v)) are the main proteins involved in the propagation of the cardiac action potential, and their phenotype affects cardiac performance. Therefore, we examined the expression of Na(v) during proliferation and differentiation in skeletal myocytes. METHODS AND RESULTS: We used the rat neonatal skeletal myocyte cell line L6E9. Proliferation of L6E9 cells induced Na(v)1.4 and Na(v)1.5, although neither protein has an apparent role in cell growth. During myogenesis, Na(v)1.5 was largely induced. Electrophysiological and pharmacological properties, as well as mRNA expression, indicate that cardiac-type Na(v)1.5 accounts for almost 90% of the Na(+) current in myotubes. Unlike in proliferation, this protein plays a pivotal role in myogenesis. The adoption of a cardiac-like phenotype is further supported by the increase in Na(v)1.5 colocalization in caveolae. Finally, we demonstrate that the treatment of myoblasts with neuregulin further increased Na(v)1.5 in skeletal myocytes. CONCLUSION: Our results indicate that skeletal myotubes adopt a cardiac-like phenotype in cell culture conditions and that the expression of Na(v)1.5 acts as an underlying molecular mechanism.


Assuntos
Cardiomioplastia/métodos , Proteínas Musculares/metabolismo , Mioblastos Esqueléticos/metabolismo , Fenótipo , Canais de Sódio/metabolismo , Potenciais de Ação/fisiologia , Animais , Biópsia , Cavéolas/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células , Células Cultivadas , Humanos , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/efeitos dos fármacos , Infarto do Miocárdio/terapia , Canal de Sódio Disparado por Voltagem NAV1.5 , Neuregulina-1/farmacologia , Técnicas de Patch-Clamp , Ratos
18.
Cancer Detect Prev ; 30(4): 375-85, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16971052

RESUMO

BACKGROUND: Potassium channels (KCh) are the most diverse and ubiquitous class of ion channels. KCh control membrane potential and contribute to nerve and cardiac action potentials and neurotransmitter release. KCh are also involved in insulin release, differentiation, activation, proliferation, apoptosis, and several other physiological functions. The aim of this review is to provide an updated overview of the KCh role during the cell growth. Their potential use as pharmacological targets in cancer therapies is also discussed. METHODS: We searched PubMed (up to 2005) and identified relevant articles. Reprints were mainly obtained by on line subscription. Additional sources were identified through cross-referencing and obtained from Library services. RESULTS: KCh are responsible for some neurological and cardiovascular diseases and for a new medical discipline, channelopathies. Their role in congenital deafness, multiple sclerosis, episodic ataxia, LQT syndrome and diabetes has been proven. Furthermore, a large body of information suggests that KCh play a role in the cell cycle progression, and it is now accepted that cells require KCh to proliferate. Thus, KCh expression has been studied in a number of tumours and cancer cells. CONCLUSIONS: Cancer is far from being considered a channelopathy. However, it seems appropriate to take into account the involvement of KCh in cancer progression and pathology when developing new strategies for cancer therapy.


Assuntos
Neoplasias/terapia , Canais de Potássio/metabolismo , Animais , Proliferação de Células , Humanos , Ativação do Canal Iônico , Neoplasias/metabolismo
19.
J Biol Chem ; 281(49): 37675-85, 2006 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-17038323

RESUMO

Voltage-dependent K(+) (Kv) currents in macrophages are mainly mediated by Kv1.3, but biophysical properties indicate that the channel composition could be different from that of T-lymphocytes. K(+) currents in mouse bone marrow-derived and Raw-264.7 macrophages are sensitive to Kv1.3 blockers, but unlike T-cells, macrophages express Kv1.5. Because Shaker subunits (Kv1) may form heterotetrameric complexes, we investigated whether Kv1.5 has a function in Kv currents in macrophages. Kv1.3 and Kv1.5 co-localize at the membrane, and half-activation voltages and pharmacology indicate that K(+) currents may be accounted for by various Kv complexes in macrophages. Co-expression of Kv1.3 and Kv1.5 in human embryonic kidney 293 cells showed that the presence of Kv1.5 leads to a positive shift in K(+) current half-activation voltages and that, like Kv1.3, Kv1.3/Kv1.5 heteromers are sensitive to r-margatoxin. In addition, both proteins co-immunoprecipitate and co-localize. Fluorescence resonance energy transfer studies further demonstrated that Kv1.5 and Kv1.3 form heterotetramers. Electrophysiological and pharmacological studies of different ratios of Kv1.3 and Kv1.5 co-expressed in Xenopus oocytes suggest that various hybrids might be responsible for K(+) currents in macrophages. Tumor necrosis factor-alpha-induced activation of macrophages increased Kv1.3 with no changes in Kv.1.5, which is consistent with a hyperpolarized shift in half-activation voltage and a lower IC(50) for margatoxin. Taken together, our results demonstrate that Kv1.5 co-associates with Kv1.3, generating functional heterotetramers in macrophages. Changes in the oligomeric composition of functional Kv channels would give rise to different biophysical and pharmacological properties, which could determine specific cellular responses.


Assuntos
Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Macrófagos/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Feminino , Humanos , Técnicas In Vitro , Canal de Potássio Kv1.3/química , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.5/química , Canal de Potássio Kv1.5/genética , Macrófagos/ultraestrutura , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Imunoeletrônica , Oócitos/metabolismo , Estrutura Quaternária de Proteína , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA