Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Int J Mol Sci ; 23(15)2022 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-35955591

RESUMO

Carcinogenesis is a multistage process involving the dysregulation of multiple genes, proteins, and pathways that make any normal cell acquire a cancer cell phenotype. Therefore, it is no surprise that numerous ion channels could be involved in this process. Since their discovery and subsequent cloning, ion channels have been established as therapeutic targets in excitable cell pathologies (e.g., cardiac arrhythmias or epilepsy); however, their involvement in non-excitable cell pathologies is relatively recent. Among all ion channels, the voltage-gated potassium channels Kv10.1 have been established as a promising target in cancer treatment due to their high expression in tumoral tissues compared to low levels in healthy tissues.


Assuntos
Epilepsia , Neoplasias , Carcinogênese/metabolismo , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Neoplasias/genética
2.
Methods Mol Biol ; 2796: 97-103, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38856897

RESUMO

The development of cell-based fluorescent assays has resulted in an incredible tool for searching new ion channels' modulators with a biophysical and clinical profile. Among all the ion channels, potassium (K+)-permeable channels represent the most diverse and relevant for cell function, making them attractive targets for drug discovery. Some of the cell-based assays for K+ channels take advantage of a thallium-sensitive dye whose fluorescence increased upon the binding of thallium (Tl+), an ion able to move through K+ channels. We optimize the FLIPR Potassium Assay Kit based on thallium influx to measure the Kv10.1 activity.


Assuntos
Tálio , Tálio/metabolismo , Humanos , Corantes Fluorescentes/química , Células HEK293 , Fluorescência , Canais de Potássio Éter-A-Go-Go
3.
Front Pharmacol ; 14: 1238503, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37554982

RESUMO

Resting membrane potential is a bioelectric property of all cells. Multiple players govern this property, the ion channels being the most important. Ion channel dysfunction can affect cells' resting membrane potential and could be associated with numerous diseases. Therefore, the drug discovery focus on ion channels has increased yearly. In addition to patch-clamp, cell-based fluorescent assays have shown a rapid and reliable method for searching new ion channel modulators. Here, we used a cell-based membrane potential assay to search for new blockers of the Kv10.1, a potassium channel strongly associated with cancer progression and a promising target in anticancer therapy. We found that fluoxetine and miconazole can inhibit the Kv10.1 channel in the micromolar range. In contrast, BL-1249 potentiates Kv10.1 currents in a dose-dependent manner, becoming the first molecule described as an activator of the channel. These results demonstrate that cell-based membrane potential assay can accelerate the discovery of new Kv10.1 modulators.

4.
Toxicon X ; 18: 100151, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36873112

RESUMO

Venoms from tarantulas contain low molecular weight vasodilatory compounds whose biological action is conceived as part of the envenomation strategy due to its propagative effects. However, some properties of venom-induced vasodilation do not match those described by such compounds, suggesting that other toxins may cooperate with these ones to produce the observed biological effect. Owing to the distribution and function of voltage-gated ion channels in blood vessels, disulfide-rich peptides isolated from venoms of tarantulas could be conceived into potential vasodilatory compounds. However, only two peptides isolated from spider venoms have been investigated so far. This study describes for the first time a subfraction containing inhibitor cystine knot peptides, PrFr-I, obtained from the venom of the tarantula Poecilotheria regalis. This subfraction induced sustained vasodilation in rat aortic rings independent of vascular endothelium and endothelial ion channels. Furthermore, PrFr-I decreased calcium-induced contraction of rat aortic segments and reduced extracellular calcium influx to chromaffin cells by the blockade of L-type voltage-gated calcium channels. This mechanism was unrelated to the activation of potassium channels from vascular smooth muscle, since vasodilation was not affected in the presence of TEA, and PrFr-I did not modify the conductance of the voltage-gated potassium channel Kv10.1. This work proposes a new envenomating function of peptides from venoms of tarantulas, and establishes a new mechanism for venom-induced vasodilation.

5.
J Neurophysiol ; 108(8): 2264-75, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22832570

RESUMO

Properties, developmental regulation, and cAMP modulation of the hyperpolarization-activated current (I(h)) were investigated by the whole cell patch-clamp technique in vestibular ganglion neurons of the rat at two postnatal stages (P7-10 and P25-28). In addition, by RT-PCR and immunohistochemistry the identity and distribution of hyperpolarization-activated and cyclic nucleotide-gated channel (HCN) isoforms that generate I(h) were investigated. I(h) current density was larger in P25-28 than P7-10 rats, increasing 410% for small cells (<30 pF) and 200% for larger cells (>30 pF). The half-maximum activation voltage (V(1/2)) of I(h) was -102 mV in P7-10 rats and in P25-28 rats shifted 7 mV toward positive voltages. At both ages, intracellular cAMP increased I(h) current density, decreased its activation time constant (τ), and resulted in a rightward shift of V(1/2) by 9 mV. Perfusion of 8-BrcAMP increased I(h) amplitude and speed up its activation kinetics. I(h) was blocked by Cs(+), zatebradine, and ZD7288. As expected, these drugs also reduced the voltage sag caused with hyperpolarizing pulses and prevented the postpulse action potential generation without changes in the resting potential. RT-PCR analysis showed that HCN1 and HCN2 subunits were predominantly amplified in vestibular ganglia and end organs and HCN3 and HCN4 to a lesser extent. Immunohistochemistry showed that the four HCN subunits were differentially expressed (HCN1 > HCN2 > HCN3 ≥ HCN4) in ganglion slices and in cultured neurons at both P7-10 and P25-28 stages. Developmental changes shifted V(1/2) of I(h) closer to the resting membrane potential, increasing its functional role. Modulation of I(h) by cAMP-mediated signaling pathway constitutes a potentially relevant control mechanism for the modulation of afferent neuron discharge.


Assuntos
Potenciais de Ação/fisiologia , AMP Cíclico/metabolismo , Neurônios/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Subunidades Proteicas/fisiologia , Ratos , Ratos Long-Evans , Núcleos Vestibulares/citologia , Núcleos Vestibulares/crescimento & desenvolvimento
6.
Appl Radiat Isot ; 187: 110331, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35764005

RESUMO

Exposure of biological systems to a radiation absorbed dose produces early and late radiogenic responses, such as ion channel modulation, oxidative stress, cell migration enhancement, and metabolic changes that could impact the efficiency of radiotherapy. To understand how radiation modulates ion channels, we irradiated HEK cells stably expressing the human ether à-go-go potassium channel-1 with gamma photons in the dose range of 2-10 Gy (60Co, 0.2 Gy/min) and measured ionic currents generated by the channel. The importance of the Kv10.1 modulation by gamma radiation was studied using cell proliferation. Results showed that a radiation-absorbed dose of 4 Gy significantly reduced the Kv10.1-evoked currents by depolarizing pulses between -100 mV and +50 mV. Additionally, the expression of Kv10.1 positively modulates HEK293 proliferation and, certainly, prevents the effect of gamma radiation on this phenomenon. Results allow concluding that the modulation of the functional expression of the Kv10.1 channel, induced by gamma radiation, leads to the expression of a radioresistant phenotype in Kv10.1 expressing cells.


Assuntos
Raios gama , Proliferação de Células , Células HEK293 , Humanos
7.
Pharmacol Rep ; 73(6): 1744-1753, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34213738

RESUMO

BACKGROUND: Ion channels have been proposed as therapeutic targets for different types of malignancies. One of the most studied ion channels in cancer is the voltage-gated potassium channel ether-à-go-go 1 or Kv10.1. Various studies have shown that Kv10.1 expression induces the proliferation of several cancer cell lines and in vivo tumor models, while blocking or silencing inhibits proliferation. Kv10.1 is a promising target for drug discovery modulators that could be used in cancer treatment. This work aimed to screen for new Kv10.1 channel modulators using a thallium influx-based assay. METHODS: Pharmacological effects of small molecules on Kv10.1 channel activity were studied using a thallium-based fluorescent assay and patch-clamp electrophysiological recordings, both performed in HEK293 stably expressing the human Kv10.1 potassium channel. RESULTS: In thallium-sensitive fluorescent assays, we found that the small molecules loperamide and amitriptyline exert a potent inhibition on the activity of the oncogenic potassium channel Kv10.1. These results were confirmed by electrophysiological recordings, which showed that loperamide and amitriptyline decreased the amplitude of Kv10.1 currents in a dose-dependent manner. Both drugs could be promising tools for further studies. CONCLUSIONS: Thallium-sensitive fluorescent assay represents a reliable methodological tool for the primary screening of different molecules with potential activity on Kv10.1 channels or other K+ channels.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Loperamida/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Relação Dose-Resposta a Droga , Fluorescência , Células HEK293 , Humanos , Loperamida/administração & dosagem , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/administração & dosagem , Reprodutibilidade dos Testes , Tálio/metabolismo
8.
Neurosci Lett ; 736: 135267, 2020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-32717335

RESUMO

Nociceptive stimuli attributes are codified in the periphery; at this level, D2-like dopamine (DA) receptor activation decreases the high voltage-gated Ca2+ current predominantly in mechanonociceptive neurons, which explains the presynaptic action mechanism of the antinociception produced by quinpirole when it is intrathecally administered in rats. However, the identity of D2-like DA receptor subtype that mediates this effect remains unknown. To answer this question, we used Fluo-4-based Ca2+ microfluorometry to study the depolarization-elicited [Ca2+]i increase in small non-peptidergic DRG neurons (identified by its binding to the Isolectin B4), and to test the effect of D2-like DA receptor activation by quinpirole in presence of selective antagonists for D2, D3, and D4 DA receptors. The results showed a significantly greater contribution of the D4 DA receptor in the down-modulation of depolarization-elicited [Ca2+]i increase in small non-peptidergic DRG neurons compared to the other receptors. Although the D2 and D3 receptor antagonists also slightly inhibited the effect of quinpirole, their effects were significantly weaker than those of the D4 receptor antagonist. Furthermore, we showed that quinpirole selectively inhibits the CaV2.2 Ca2+ channels. Our results suggest that the activation of the D4 DA receptors is a promising strategy for pain management at the spinal cord level.


Assuntos
Canais de Cálcio Tipo N/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Neurônios/metabolismo , Quimpirol/farmacologia , Receptores de Dopamina D4/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo N/metabolismo , Células Cultivadas , Feminino , Gânglios Espinais/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar
9.
Acta Histochem ; 121(8): 151440, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31561916

RESUMO

Thyroid hormones (THs) regulate several physiological processes in female mammals, many of which are related to reproduction such as steroidogenesis in the ovary, oocyte and granulosa cells maturation, follicular development and differentiation, and ovulation. THs actions require the presence of THs transporters to facilitate their cellular uptake and efflux. MCT8 and OATP1C1 are the principal THs transporters. The aim of the present study was to determine the gene expression and cellular localization of MCT8 and OATP1C1 in the rat ovary during the diestrus-II cycle phase. Ovaries of virgin adult rats were histologically processed. Reverse Transcription-PCR and immunohistochemistry analyses for MCT8 and OATP1C1 were done. MCT8 gene expression level was significantly higher (P ≤ 0.01) than that of OATP1C1 in the rat ovary. MCT8 and OATP1C1 were found in all types of ovarian cells but with different immunoreactivity. MCT8 showed stronger immunoreactivity in tertiary and Graafian follicles, corpus luteum and blood vessels, whereas OATP1C1's immunoreactivity was stronger in stroma cells, tunica albuginea, and blood vessels. Our results provide evidence that THs and their transporters are both necessary for ovarian function and that any alteration in these transporters could interfere with reproductive processes such as ovulation and steroidogenesis, compromising fertility.


Assuntos
Regulação da Expressão Gênica/fisiologia , Transportadores de Ácidos Monocarboxílicos/biossíntese , Proteínas de Transporte de Cátions Orgânicos/biossíntese , Ovário/metabolismo , Animais , Feminino , Imuno-Histoquímica , Ovário/citologia , Ratos , Ratos Wistar
10.
Neuroscience ; 417: 81-94, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31430528

RESUMO

Intrathecal (i.t.) administration of quinpirole, a dopamine (DA) D2-like receptor agonist, produces antinociception to mechanonociceptive stimuli but not to thermonociceptive stimuli. To determine a cellular mechanism for the specific antinociceptive effect of D2-like receptor activation on mechanonociception, we evaluated the effect of quinpirole on voltage-gated Ca2+ influx in cultured dorsal root ganglion (DRG) neurons and the D2 DA receptor distribution in subpopulations of rat nociceptive DRG neurons. Small-diameter DRG neurons were classified into IB4+ (nonpeptidergic) and IB4- (peptidergic). Intracellular [Ca2+] microfluorometry and voltage-clamp experiments showed that quinpirole reduced Ca2+ influx and inhibited the high voltage-activated Ca2+ current (HVA-ICa) in half of IB4+ neurons, leaving Ca2+ entry and HVA-ICa in IB4- neurons nearly unaffected. Pretreatment with ω-conotoxin MVIIA prevented the effect of quinpirole on HVA-ICa from IB4+ neurons, indicating that quinpirole mainly inhibits CaV2.2 channels. Immunofluorescence experiments showed that D2 DA receptor was present mainly in IB4+ small DRG neurons. Finally, in behavioral experiments in rats, the clinically approved D2-like receptor agonist pramipexole produced spinal antinociception in a similar fashion to quinpirole, with a significant effect only in the mechanonociceptive test. Our results explain, at least in part, why D2-like receptor agonists produce antinociception on mechanonociceptors.


Assuntos
Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Receptores de Dopamina D2/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiologia , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/farmacologia , Animais , Cálcio/metabolismo , Cálcio/fisiologia , Agonistas de Dopamina/farmacologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiologia , Masculino , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Nociceptores/fisiologia , Pramipexol/farmacologia , Quimpirol/farmacologia , Ratos , Ratos Wistar , Medula Espinal/metabolismo
11.
Stem Cells Int ; 2019: 7627148, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31065279

RESUMO

Human mesenchymal stem cells (MSCs) are good candidates for brain cell replacement strategies and have already been used as adjuvant treatments in neurological disorders. MSCs can be obtained from many different sources, and the present study compares the potential of neuronal transdifferentiation in MSCs from adult and neonatal sources (Wharton's jelly (WhJ), dental pulp (DP), periodontal ligament (PDL), gingival tissue (GT), dermis (SK), placenta (PLAC), and umbilical cord blood (UCB)) with a protocol previously tested in bone marrow- (BM-) MSCs consisting of a cocktail of six small molecules: I-BET151, CHIR99021, forskolin, RepSox, Y-27632, and dbcAMP (ICFRYA). Neuronal morphology and the presence of cells positive for neuronal markers (TUJ1 and MAP2) were considered attributes of neuronal induction. The ICFRYA cocktail did not induce neuronal features in WhJ-MSCs, and these features were only partial in the MSCs from dental tissues, SK-MSCs, and PLAC-MSCs. The best response was found in UCB-MSCs, which was comparable to the response of BM-MSCs. The addition of neurotrophic factors to the ICFRYA cocktail significantly increased the number of cells with complex neuron-like morphology and increased the number of cells positive for mature neuronal markers in BM- and UCB-MSCs. The neuronal cells generated from UCB-MSCs and BM-MSCs showed increased reactivity of the neuronal genes TUJ1, MAP2, NF-H, NCAM, ND1, TAU, ENO2, GABA, and NeuN as well as down- and upregulation of MSC and neuronal genes, respectively. The present study showed marked differences between the MSCs from different sources in response to the transdifferentiation protocol used here. These results may contribute to identifying the best source of MSCs for potential cell replacement therapies.

12.
Nat Commun ; 6: 8095, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26311398

RESUMO

Hyaluronan (HA) is present in the extracellular matrix of all body tissues, including synovial fluid in joints, in which it behaves as a filter that buffers transmission of mechanical forces to nociceptor nerve endings thereby reducing pain. Using recombinant systems, mouse-cultured dorsal root ganglia (DRG) neurons and in vivo experiments, we found that HA also modulates polymodal transient receptor potential vanilloid subtype 1 (TRPV1) channels. HA diminishes heat, pH and capsaicin (CAP) responses, thus reducing the opening probability of the channel by stabilizing its closed state. Accordingly, in DRG neurons, HA decreases TRPV1-mediated impulse firing and channel sensitization by bradykinin. Moreover, subcutaneous HA injection in mice reduces heat and capsaicin nocifensive responses, whereas the intra-articular injection of HA in rats decreases capsaicin joint nociceptor fibres discharge. Collectively, these results indicate that extracellular HA reduces the excitability of the ubiquitous TRPV1 channel, thereby lowering impulse activity in the peripheral nociceptor endings underlying pain.


Assuntos
Adjuvantes Imunológicos/farmacologia , Ácido Hialurônico/farmacologia , Neurônios/efeitos dos fármacos , Dor Nociceptiva , Nociceptores/efeitos dos fármacos , Joelho de Quadrúpedes/efeitos dos fármacos , Canais de Cátion TRPV/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Bradicinina/farmacologia , Células CHO , Cálcio/metabolismo , Capsaicina/farmacologia , Linhagem Celular Tumoral , Cricetulus , Gânglios Espinais/citologia , Células HEK293 , Temperatura Alta , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Modelos Moleculares , Mutagênese Sítio-Dirigida , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Fármacos do Sistema Sensorial/farmacologia , Joelho de Quadrúpedes/inervação , Canal de Cátion TRPA1 , Canais de Cátion TRPM/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/metabolismo , Vasodilatadores/farmacologia
13.
Cell Rep ; 8(5): 1571-82, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25199828

RESUMO

Animals sense cold ambient temperatures through the activation of peripheral thermoreceptors that express TRPM8, a cold- and menthol-activated ion channel. These receptors can discriminate a very wide range of temperatures from innocuous to noxious. The molecular mechanism responsible for the variable sensitivity of individual cold receptors to temperature is unclear. To address this question, we performed a detailed ion channel expression analysis of cold-sensitive neurons, combining bacterial artificial chromosome (BAC) transgenesis with a molecular-profiling approach in fluorescence-activated cell sorting (FACS)-purified TRPM8 neurons. We found that TASK-3 leak potassium channels are highly enriched in a subpopulation of these sensory neurons. The thermal threshold of TRPM8 cold neurons is decreased during TASK-3 blockade and in mice lacking TASK-3, and, most importantly, these mice display hypersensitivity to cold. Our results demonstrate a role of TASK-3 channels in thermosensation, showing that a channel-based combinatorial strategy in TRPM8 cold thermoreceptors leads to molecular specialization and functional diversity.


Assuntos
Canais de Potássio/metabolismo , Canais de Cátion TRPM/metabolismo , Sensação Térmica , Animais , Células Cultivadas , Temperatura Baixa , Camundongos , Células do Corno Posterior/metabolismo , Células do Corno Posterior/fisiologia , Canais de Potássio/genética , Limiar Sensorial , Canais de Cátion TRPM/genética
14.
Nat Commun ; 5: 3125, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24445575

RESUMO

Gram-negative bacterial infections are accompanied by inflammation and somatic or visceral pain. These symptoms are generally attributed to sensitization of nociceptors by inflammatory mediators released by immune cells. Nociceptor sensitization during inflammation occurs through activation of the Toll-like receptor 4 (TLR4) signalling pathway by lipopolysaccharide (LPS), a toxic by-product of bacterial lysis. Here we show that LPS exerts fast, membrane delimited, excitatory actions via TRPA1, a transient receptor potential cation channel that is critical for transducing environmental irritant stimuli into nociceptor activity. Moreover, we find that pain and acute vascular reactions, including neurogenic inflammation (CGRP release) caused by LPS are primarily dependent on TRPA1 channel activation in nociceptive sensory neurons, and develop independently of TLR4 activation. The identification of TRPA1 as a molecular determinant of direct LPS effects on nociceptors offers new insights into the pathogenesis of pain and neurovascular responses during bacterial infections and opens novel avenues for their treatment.


Assuntos
Lipopolissacarídeos/efeitos adversos , Inflamação Neurogênica/metabolismo , Dor/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Escherichia coli/química , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Lipídeo A/química , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inflamação Neurogênica/patologia , Neuropeptídeos/metabolismo , Nociceptores/metabolismo , Dor/patologia , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Canal de Cátion TRPA1 , Receptor 4 Toll-Like/metabolismo , Canais de Potencial de Receptor Transitório/agonistas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA