Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.368
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
J Biol Chem ; 297(4): 101126, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34461097

RESUMO

Transient receptor potential (TRP) cation channels, which are conserved across mammals, flies, fish, sea squirts, worms, and fungi, essentially contribute to cellular Ca2+ signaling. The activity of the unique TRP channel in yeast, TRP yeast channel 1 (TRPY1), relies on the vacuolar and cytoplasmic Ca2+ concentration. However, the mechanism(s) of Ca2+-dependent regulation of TRPY1 and possible contribution(s) of Ca2+-binding proteins are yet not well understood. Our results demonstrate a Ca2+-dependent binding of yeast calmodulin (CaM) to TRPY1. TRPY1 activity was increased in the cmd1-6 yeast strain, carrying a non-Ca2+-binding CaM mutant, compared with the parent strain expressing wt CaM (Cmd1). Expression of Cmd1 in cmd1-6 yeast rescued the wt phenotype. In addition, in human embryonic kidney 293 cells, hypertonic shock-induced TRPY1-dependent Ca2+ influx and Ca2+ release were increased by the CaM antagonist ophiobolin A. We found that coexpression of mammalian CaM impeded the activity of TRPY1 by reinforcing effects of endogenous CaM. Finally, inhibition of TRPY1 by Ca2+-CaM required the cytoplasmic amino acid stretch E33-Y92. In summary, our results show that TRPY1 is under inhibitory control of Ca2+-CaM and that mammalian CaM can replace yeast CaM for this inhibition. These findings add TRPY1 to the innumerable cellular proteins, which include a variety of ion channels, that use CaM as a constitutive or dissociable Ca2+-sensing subunit, and contribute to a better understanding of the modulatory mechanisms of Ca2+-CaM.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Calmodulina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Canais de Cátion TRPC/metabolismo , Vacúolos/metabolismo , Cálcio/química , Calmodulina/antagonistas & inibidores , Calmodulina/química , Calmodulina/genética , Células HEK293 , Humanos , Domínios Proteicos , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Sesterterpenos/farmacologia , Canais de Cátion TRPC/química , Canais de Cátion TRPC/genética , Vacúolos/química , Vacúolos/genética
2.
Mol Pharmacol ; 100(3): 237-257, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34127538

RESUMO

Ion channels are attractive drug targets for many therapeutic applications. However, high-throughput screening (HTS) of drug candidates is difficult and remains very expensive. We thus assessed the suitability of the bioluminescence resonance energy transfer (BRET) technique as a new HTS method for ion-channel studies by taking advantage of our recently characterized intra- and intermolecular BRET probes targeting the transient receptor potential vanilloid type 1 (TRPV1) ion channel. These BRET probes monitor conformational changes during TRPV1 gating and subsequent coupling with calmodulin, two molecular events that are intractable using reference techniques such as automated calcium assay (ACA) and automated patch-clamp (APC). We screened the small-sized Prestwick chemical library, encompassing 1200 compounds with high structural diversity, using either intra- and intermolecular BRET probes or ACA. Secondary screening of the detected hits was done using APC. Multiparametric analysis of our results shed light on the capability of calmodulin inhibitors included in the Prestwick library to inhibit TRPV1 activation by capsaicin. BRET was the lead technique for this identification process. Finally, we present data exemplifying the use of intramolecular BRET probes to study other transient receptor potential (TRP) channels and non-TRPs ion channels. Knowing the ease of use of BRET biosensors and the low cost of the BRET technique, these assays may advantageously be included for extending ion-channel drug screening. SIGNIFICANCE STATEMENT: This study screened a chemical library against TRPV1 ion channel using bioluminescence resonance energy transfer (BRET) molecular probes and compared the results with the ones obtained using reference techniques such as automated calcium assay and automated patch-clamp. Multiparametric analysis of our results shed light on the capability of calmodulin antagonists to inhibit chemical activation of TRPV1 and indicates that BRET probes may advantageously be included in ion channel drug screening campaigns.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Canais de Cátion TRPV/metabolismo , Bioensaio/métodos , Cálcio/química , Calmodulina/antagonistas & inibidores , Células HEK293 , Humanos , Ligantes , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Bibliotecas de Moléculas Pequenas , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/antagonistas & inibidores
3.
J Cell Sci ; 131(2)2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-28183730

RESUMO

Ca2+ is a second messenger in many physiological and phytopathological processes. Peroxisomes are subcellular compartments with an active oxidative and nitrosative metabolism. Previous studies have demonstrated that peroxisomal nitric oxide (NO) generation is dependent on Ca2+ and calmodulin (CaM). Here, we used Arabidopsis thaliana transgenic seedlings expressing cyan fluorescent protein (CFP) containing a type 1 peroxisomal-targeting signal motif (PTS1; CFP-PTS1), which enables peroxisomes to be visualized in vivo, and also used a cell-permeable fluorescent probe for Ca2+ Analysis by confocal laser-scanning microscopy (CLSM) enabled us to visualize the presence of endogenous Ca2+ in the peroxisomes of both roots and guard cells. The presence of Ca2+ in peroxisomes and the import of CFP-PTS1 are drastically disrupted by both CaM antagonist and glutathione (GSH). Furthermore, the activity of three peroxisomal enzymes (catalase, glycolate oxidase and hydroxypyruvate reductase) containing PTS1 was clearly affected in these conditions, with a decrease of between 41 and 51%. In summary, data show that Ca2+ and CaM are strictly necessary for protein import and normal functionality of peroxisomal enzymes, including antioxidant and photorespiratory enzymes, as well as for NO production.


Assuntos
Arabidopsis/metabolismo , Cálcio/metabolismo , Calmodulina/antagonistas & inibidores , Peroxissomos/metabolismo , Arabidopsis/citologia , Arabidopsis/efeitos dos fármacos , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Calmodulina/metabolismo , Cloroplastos/efeitos dos fármacos , Cloroplastos/metabolismo , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Glutationa/farmacologia , Óxido Nítrico/farmacologia , Peroxissomos/efeitos dos fármacos , Células Vegetais/efeitos dos fármacos , Células Vegetais/metabolismo , Raízes de Plantas/citologia , Raízes de Plantas/efeitos dos fármacos , Raízes de Plantas/metabolismo , Transporte Proteico/efeitos dos fármacos
4.
Anal Biochem ; 572: 25-32, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30825429

RESUMO

The KRAS gene is highly mutated in human cancers and the focus of current Ras drug development efforts. Recently the interface between the C-terminus of K-Ras and calmodulin (CaM) was proposed as a target site to block K-Ras driven cancer cell stemness. We therefore aimed at developing a high-throughput amenable screening assay to identify novel CaM-inhibitors as potential K-Ras stemness-signaling disruptors. A modulated time-resolved Förster resonance energy transfer (mTR-FRET)-assay was developed and benchmarked against an identically designed fluorescence anisotropy (FA)-assay. In both assays, two CaM-binding peptides were labeled with Eu(III)-chelate or fluorescein and used as single-label reporter probes that were displaced from CaM upon competitor binding. Thus, peptidic and small molecule competitors with nanomolar to micromolar affinities to CaM could be detected, including a peptide that was derived from the C-terminus of K-Ras. In order to detect CaM-residue specific covalent inhibitors, a cell lysate-based Förster resonance energy transfer (FRET)-assay was furthermore established. This assay enabled us to measure the slow, residue-specific, covalent inhibition by ophiobolin A in the presence of other endogenous proteins. In conclusion, we have developed a panel of fluorescence-assays that allows identification of conventional and covalent CaM-inhibitors as potential disruptors of K-Ras driven cancer cell stemness.


Assuntos
Calmodulina/antagonistas & inibidores , Inibidores Enzimáticos/química , Transferência Ressonante de Energia de Fluorescência/métodos , Ensaios de Triagem em Larga Escala/métodos , Calmodulina/genética , Calmodulina/metabolismo , Inibidores Enzimáticos/metabolismo , Európio/química , Fluoresceína/química , Humanos , Concentração Inibidora 50 , Cinética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Sesterterpenos/química , Sesterterpenos/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo
5.
J Cell Biochem ; 119(7): 6216-6230, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29663486

RESUMO

Patients with triple negative breast cancer (TNBC) have no successful "targeted" treatment modality, which represents a priority for novel therapy strategies. Upregulated death receptor 5 (DR5) expression levels in breast cancer cells compared to normal cells enable TRA-8, a DR5 specific agonistic antibody, to specifically target malignant cells for apoptosis without inducing normal hepatocyte apoptosis. Drug resistance is a common obstacle in TRAIL-based therapy for TNBC. Calmodulin (CaM) is overexpressed in breast cancer. In this study, we characterized the novel function of CaM antagonist in enhancing TRA-8 induced cytotoxicity in TRA-8 resistant TNBC cells and its underlying molecular mechanisms. Results demonstrated that CaM antagonist(s) enhanced TRA-8 induced cytotoxicity in a concentration and time-dependent manner for TRA-8 resistant TNBC cells. CaM directly bound to DR5 in a Ca2+ dependent manner, and CaM siRNA promoted DR5 recruitment of FADD and caspase-8 for DISC formation and TRA-8 activated caspase cleavage for apoptosis in TRA-8 resistant TNBC cells. CaM antagonist, trifluoperazine, enhanced TRA-8 activated DR5 oligomerization, DR5-mediated DISC formation, and TRA-8 activated caspase cleavage for apoptosis, and decreased anti-apoptotic pERK, pAKT, XIAP, and cIAP-1 expression in TRA-8 resistant TNBC cells. These results suggest that CaM could be a key regulator to mediate DR5-mediated apoptotic signaling, and suggests a potential strategy for using CaM antagonists to overcome drug resistance of TRAIL-based therapy for TRA-8 resistant TNBC.


Assuntos
Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Calmodulina/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Feminino , Humanos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Células Tumorais Cultivadas
6.
J Neurochem ; 144(1): 68-80, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29063596

RESUMO

Gq/11 protein-coupled human histamine H1 receptors in Chinese hamster ovary cells stimulated with histamine undergo clathrin-dependent endocytosis followed by proteasome/lysosome-mediated down-regulation. In this study, we evaluated the effects of a sustained increase in intracellular Ca2+ concentrations induced by a receptor-bypassed stimulation with ionomycin, a Ca2+ ionophore, on the endocytosis and down-regulation of H1 receptors in Chinese hamster ovary cells. All cellular and cell-surface H1 receptors were detected by the binding of [3 H]mepyramine to intact cells sensitive to the hydrophobic and hydrophilic H1 receptor ligands, mepyramine and pirdonium, respectively. The pretreatment of cells with ionomycin markedly reduced the mepyramine- and pirdonium-sensitive binding sites of [3 H]mepyramine, which were completely abrogated by the deprivation of extracellular Ca2+ and partially by a ubiquitin-activating enzyme inhibitor (UBEI-41), but were not affected by inhibitors of calmodulin (W-7 or calmidazolium) and protein kinase C (chelerythrine or GF109203X). These ionomycin-induced changes were also not affected by inhibitors of receptor endocytosis via clathrin (hypertonic sucrose) and caveolae/lipid rafts (filipin or nystatin) or by inhibitors of lysosomes (E-64, leupeptin, chloroquine, or NH4 Cl), proteasomes (lactacystin or MG-132), and a Ca2+ -dependent non-lysosomal cysteine protease (calpain) (MDL28170). Since H1 receptors were normally detected by confocal immunofluorescence microscopy with an antibody against H1 receptors, even after the ionomycin treatment, H1 receptors appeared to exist in a form to which [3 H]mepyramine was unable to bind. These results suggest that H1 receptors are apparently down-regulated by a sustained increase in intracellular Ca2+ concentrations with no process of endocytosis and lysosomal/proteasomal degradation of receptors.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/farmacologia , Receptores Histamínicos H1/biossíntese , Animais , Astrocitoma , Células CHO , Ionóforos de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Calmodulina/antagonistas & inibidores , Calpaína/antagonistas & inibidores , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Regulação para Baixo/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Histamina/farmacologia , Humanos , Fosfatos de Inositol/metabolismo , Ionomicina/farmacologia , Lisossomos/efeitos dos fármacos , Microdomínios da Membrana/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Pirilamina/metabolismo , Receptores Histamínicos H1/genética , Proteínas Recombinantes/biossíntese
7.
Biochim Biophys Acta Proteins Proteom ; 1866(5-6): 617-623, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29555454

RESUMO

We compare the binding of an inhibitor with that of a natural peptide to Ca2+ saturated calmodulin (holo-CaM). As inhibitor we have chosen trifluoperazine (TFP) that is inducing a huge conformational change of holo-CaM from the open dumbbell-shaped to the closed globular conformation upon binding. On the other hand, melittin is used as model peptide, which is a well-known natural binding partner of holo-CaM. The experiments are carried out as a function of pressure to reveal the contribution of volume or packing effects to the stability of the calmodulin-ligand complexes. From high-pressure Fourier transform infrared (FTIR) spectroscopy, we find that the holo-CaM/TFP complex has a much higher pressure stability than the holo-CaM/melittin complex. Although the analysis of the secondary structure of holo-CaM (without and with ligand) indicates no major changes up to several kbar, pressure-induced exposure of α-helices to water is most pronounced for holo-CaM without ligand, followed by holo-CaM/melittin and then holo-CaM/TFP. Moreover, structural pressure resistance of the holo-CaM/TFP complex in comparison with the holo-CaM/melittin complex is also clearly visible by higher Ca2+ affinity. Förster resonance energy transfer (FRET) from the Tyr residues of holo-CaM to the Trp residue of melittin even suggests some partial dissociation of the complex under pressure which points to void volumes at the protein-ligand interface and to electrostatic binding. Thus, all results of this study show that the inhibitor TFP binds to holo-CaM with higher packing density than the peptide melittin enabling a favorable volume contribution to the inhibitor efficiency.


Assuntos
Calmodulina/metabolismo , Transferência Ressonante de Energia de Fluorescência , Meliteno/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Trifluoperazina/metabolismo , Animais , Sítios de Ligação , Calmodulina/antagonistas & inibidores , Calmodulina/química , Ligantes , Meliteno/química , Modelos Moleculares , Pressão , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Ratos , Relação Estrutura-Atividade , Trifluoperazina/química
8.
J Reprod Dev ; 64(5): 445-449, 2018 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-29887540

RESUMO

We aimed to test whether the calmodulin (CaM) inhibitors, calmidazolium (CZ) and N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide (W-7), can be used to assess lipid disorder by flow cytometry using Merocyanine 540 (M540). Boar spermatozoa were incubated in non-capacitating conditions for 10 min at room temperature with 1 µM CZ, 200 µM W-7, or 1 mM 8-bromoadenosine 3',5'-cyclic monophosphate (8-Br-cAMP). Then, sperm were 1) directly evaluated, 2) centrifuged and washed prior to evaluation, or 3) diluted with PBS prior to evaluation. Direct evaluation showed an increase in high M540 fluorescence in spermatozoa treated with the inhibitors (4.7 ± 1.8 [control] vs. 70.4 ± 4.0 [CZ] and 71.4 ± 4.2 [W-7], mean % ± SD, P < 0.001); washing decreased the percentage of sperm showing high M540 fluorescence for W-7 (4.8 ± 2.2, mean % ± SD) but not for CZ (69.4 ± 3.9, mean % ± SD, P < 0.001), and dilution showed an increase in high M540 fluorescence for both CZ and W-7; 8-Br-cAMP did not induce a rise in sperm showing high M540 fluorescence. Therefore, special care must be taken when M540 is used in spermatozoa with CaM inhibitors.


Assuntos
Calmodulina/antagonistas & inibidores , Pirimidinonas/química , Capacitação Espermática , Espermatozoides/efeitos dos fármacos , 8-Bromo Monofosfato de Adenosina Cíclica/química , Animais , Membrana Celular/efeitos dos fármacos , Citometria de Fluxo , Lipídeos/química , Masculino , Suínos
9.
J Cell Physiol ; 232(12): 3496-3509, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28105751

RESUMO

An upregulation of Egr-1 expression has been reported in models of atherosclerosis and intimal hyperplasia and, various vasoactive peptides and growth promoting stimuli have been shown to induce the expression of Egr-1 in vascular smooth muscle cells (VSMC). Angiotensin-II (Ang-II) is a key vasoactive peptide that has been implicated in the pathogenesis of vascular diseases. Ang-II elevates intracellular Ca2+ through activation of the store-operated calcium entry (SOCE) involving an inositol-3-phosphate receptor (IP3R)-coupled depletion of endoplasmic reticular Ca2+ and a subsequent activation of the stromal interaction molecule 1 (STIM-1)/Orai-1 complex. However, the involvement of IP3R/STIM-1/Orai-1-Ca2+ -dependent signaling in Egr-1 expression in VSMC remains unexplored. Therefore, in the present studies, we have examined the role of Ca2+ signaling in Ang-II-induced Egr-1 expression in VSMC and investigated the contribution of STIM-1 or Orai-1 in mediating this response. 2-aminoethoxydiphenyl borate (2-APB), a dual non-competitive antagonist of IP3R and inhibitor of SOCE, decreased Ang-II-induced Ca2+ release and attenuated Ang-II-induced enhanced expression of Egr-1 protein and mRNA levels. Egr-1 upregulation was also suppressed following blockade of calmodulin and CaMKII. Furthermore, RNA interference-mediated depletion of STIM-1 or Orai-1 attenuated Ang-II-induced Egr-1 expression as well as Ang-II-induced phosphorylation of ERK1/2 and CREB. In addition, siRNA-induced silencing of CREB resulted in a reduction in the expression of Egr-1 stimulated by Ang-II. In summary, our data demonstrate that Ang-II-induced Egr-1 expression is mediated by STIM-1/Orai-1/Ca2+ -dependent signaling pathways in A-10 VSMC.


Assuntos
Angiotensina II/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina/antagonistas & inibidores , Calmodulina/metabolismo , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Proteína 1 de Resposta de Crescimento Precoce/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteína ORAI1/genética , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Ratos , Molécula 1 de Interação Estromal/genética , Fatores de Tempo , Transfecção , Regulação para Cima
10.
J Cell Biochem ; 118(8): 2285-2294, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28092099

RESUMO

Activation of death receptor-5 (DR5) leads to the formation of death-inducing signaling complex (DISC) for apoptotic signaling. TRA-8, a DR5 specific agonistic antibody, has demonstrated significant cytotoxic activity in vitro and in vivo without inducing hepatotoxicity. Calmodulin (CaM) that is overexpressed in breast cancer plays a critical role in regulating DR5-mediated apoptosis. However, the mechanism of CaM in regulating DR5-mediated apoptotic signaling remains unknown. In this study, we characterized CaM binding to DR5-mediated DISC for apoptosis in TRA-8 sensitive breast cancer cell lines using co-immunoprecipitation, fluorescence microscopic imaging, caspase signaling analysis, and cell viability assay. Results show that upon DR5 activation, CaM was recruited into DR5-mediated DISC in a calcium dependent manner. CaM antagonist, trifluoperazine (TFP), inhibited CaM recruitment into the DISC and attenuated DISC formation. DR5 oligomerization is critical for DISC formation for apoptosis. TFP decreased TRA-8 activated DR5 oligomerization, which was consistent with TFP's effect on DR5-mediated DISC formation. TFP and Ca2+ chelator, EGTA, impeded TRA-8-activated caspase-dependent apoptotic signaling, and TFP decreased TRA-8-induced cell cytotoxicity. These results demonstrated CaM binding to DR5-mediated DISC in a calcium dependent manner and may identify CaM as a key regulator of DR5-mediated DISC formation for apoptosis in breast cancer. J. Cell. Biochem. 118: 2285-2294, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Calmodulina/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Neoplasias da Mama/metabolismo , Calmodulina/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Ácido Egtázico/farmacologia , Humanos , Imunoprecipitação , Ligação Proteica , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Trifluoperazina/farmacologia
11.
Eur J Neurosci ; 46(9): 2542-2547, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28925524

RESUMO

Human aquaporin 4 (AQP4) is the primary water channel protein in brain astrocytes. Hypothermia is known to cause astrocyte swelling in culture, but the precise role of AQP4 in this process is unknown. Primary human cortical astrocytes were cultured under hypothermic (32 °C) or normothermic (37 °C) conditions. AQP4 transcript, total protein and surface-localized protein were quantified using RT-qPCR, sandwich ELISA with whole cell lysates or cell surface biotinylation, followed by ELISA analysis of the surface-localized protein, respectively. Four-hour mild hypothermic treatment increased the surface localization of AQP4 in human astrocytes to 155 ± 4% of normothermic controls, despite no change in total protein expression levels. The hypothermia-mediated increase in AQP4 surface abundance on human astrocytes was blocked using either calmodulin antagonist (trifluoperazine, TFP); TRPV4 antagonist, HC-067047 or calcium chelation using EGTA-AM. The TRPV4 agonist (GSK1016790A) mimicked the effect of hypothermia compared with untreated normothermic astrocytes. Hypothermia led to an increase in surface localization of AQP4 in human astrocytes through a mechanism likely dependent on the TRPV4 calcium channel and calmodulin activation. Understanding the effects of hypothermia on astrocytic AQP4 cell surface expression may help develop new treatments for brain swelling based on an in-depth mechanistic understanding of AQP4 translocation.


Assuntos
Aquaporina 4/metabolismo , Astrócitos/metabolismo , Calmodulina/metabolismo , Córtex Cerebral/metabolismo , Hipotermia/metabolismo , Canais de Cátion TRPV/metabolismo , Aquaporina 4/antagonistas & inibidores , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Cálcio/metabolismo , Calmodulina/antagonistas & inibidores , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/patologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Humanos , Hipotermia/patologia , Hipotermia Induzida , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , RNA Mensageiro/metabolismo , Canais de Cátion TRPV/agonistas
12.
J Pharmacol Sci ; 133(3): 168-175, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28330759

RESUMO

Decreased brain glutamate level has emerged as a new therapeutic approach for epilepsy. This study investigated the effect and mechanism of amiodarone, an anti-arrhythmic drug with antiepileptic activity, on glutamate release in the rat hippocampus. In a synaptosomal preparation, amiodarone reduced 4-aminopyridine-evoked Ca2+-dependent glutamate release and cytosolic Ca2+ concentration elevation. Amiodarone did not affect the 4-aminopyridine-evoked depolarization of the synaptosomal membrane potential or the Na+ channel activator veratridine-evoked glutamate release, indicating that the amiodarone-mediated inhibition of glutamate release is not caused by a decrease in synaptosomal excitability. The inhibitory effect of amiodarone on 4-aminopyridine-evoked glutamate release was markedly decreased in synaptosomes pretreated with the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker ω-conotoxin MVIIC, the calmodulin antagonists W7 and calmidazolium, or the protein kinase A inhibitors H89 and KT5720. However, the intracellular Ca2+-release inhibitors dantrolene and CGP37157 had no effect on the amiodarone-mediated inhibition of glutamate release. Furthermore, amiodarone reduced the frequency of miniature excitatory postsynaptic currents without affecting their amplitude in hippocampal slices. Our data suggest that amiodarone reduces Ca2+ influx through N- and P/Q-type Ca2+ channels, subsequently reducing the Ca2+-calmodulin/protein kinase A cascade to inhibit the evoked glutamate release from rat hippocampal nerve terminals.


Assuntos
Amiodarona/farmacologia , Antiarrítmicos/farmacologia , Ácido Glutâmico/metabolismo , Hipocampo/efeitos dos fármacos , Sinaptossomos/efeitos dos fármacos , 4-Aminopiridina/farmacologia , Animais , Ácido Aspártico/farmacologia , Cálcio/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Calmodulina/antagonistas & inibidores , Capsaicina/farmacologia , Carbazóis/farmacologia , Hipocampo/metabolismo , Hipocampo/fisiologia , Imidazóis/farmacologia , Isoquinolinas/farmacologia , Macrolídeos/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Ratos Sprague-Dawley , Sulfonamidas/farmacologia , Sinaptossomos/metabolismo , Sinaptossomos/fisiologia , ômega-Conotoxinas/farmacologia
13.
J Neurosci Res ; 94(5): 401-8, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26864654

RESUMO

Group I metabotropic glutamate receptors (mGluR), including mGluR1 and mGluR 5 (mGluR1/5), are coupled to Gq and modulate activity-dependent synaptic plasticity. Direct activation of mGluR1/5 causes protein translation-dependent long-term depression (LTD). Although it has been established that intracellular Ca(2+) and the Gq-regulated signaling molecules are required for mGluR1/5 LTD, whether and how Ca(2+) regulates Gq signaling and upregulation of protein expression remain unknown. Through pharmacological inhibition, we tested the function of the Ca(2+) sensor calmodulin (CaM) in intracellular signaling triggered by the activation of mGluR1/5. CaM inhibitor N-[4-aminobutyl]-5-chloro-2-naphthalenesulfonamide hydrochloride (W13) suppressed the mGluR1/5-stimulated activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and p70-S6 kinase 1 (S6K1) in hippocampal neurons. W13 also blocked the mGluR1/5 agonist-induced synaptic depression in hippocampal slices and in anesthetized mice. Consistent with the function of CaM, inhibiting the downstream targets Ca(2+) /CaM-dependent protein kinases (CaMK) blocked ERK1/2 and S6K1 activation. Furthermore, disruption of the CaM-CaMK-ERK1/2 signaling cascade suppressed the mGluR1/5-stimulated upregulation of Arc expression. Altogether, our data suggest CaM as a new Gq signaling component for coupling Ca(2+) and protein upregulation and regulating mGluR1/5-mediated synaptic modification.


Assuntos
Calmodulina/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Transdução de Sinais/fisiologia , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Calmodulina/antagonistas & inibidores , Células Cultivadas , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
14.
J Exp Bot ; 67(3): 809-19, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26608645

RESUMO

Calcium (Ca(2+)) mobilization is a central theme in various plant signal transduction pathways. We demonstrate that Arabidopsis thaliana cyclic nucleotide-gated channel 2 (AtCNGC2) is involved in jasmonic acid (JA)-induced apoplastic Ca(2+) influx in Arabidopsis epidermal cells. Ca(2+) imaging results showed that JA can induce an elevation in the cytosolic cAMP concentration ([cAMP]cyt), reaching a maximum within 3 min. Dibutyryl cAMP (db-cAMP), a cell membrane-permeable analogue of cAMP, induced an increase in the cytosolic Ca(2+) concentration ([Ca(2+)]cyt), with a peak at 4 min. This [Ca(2+)]cyt increase was triggered by the JA-induced increase in [cAMP]cyt. W-7[N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide], an antagonist of calmodulin, positively modulated the JA-induced increase in [Ca(2+)]cyt, while W-5[N-(6-aminohexyl)-1-naphthalenesulfonamide], an inactive antagonist of calmodulin, had no apparent effect. db-cAMP and JA positively induced the expression of primary (i.e. JAZ1 and MYC2) and secondary (i.e. VSP1) response genes in the JA signalling pathway in wild-type Arabidopsis thaliana, whereas they had no significant effect in the AtCNGC2 mutant 'defense, no death (dnd1) plants. These data provide evidence that JA first induces the elevation of cAMP, and cAMP, as an activating ligand, activates the AtCNGC2 channel, resulting in apoplastic Ca(2+) influx through AtCNGC2.


Assuntos
Arabidopsis/metabolismo , Cálcio/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Ciclopentanos/farmacologia , Oxilipinas/farmacologia , Arabidopsis/efeitos dos fármacos , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Bucladesina/farmacologia , Calmodulina/antagonistas & inibidores , Calmodulina/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Modelos Biológicos , Folhas de Planta/efeitos dos fármacos , Folhas de Planta/metabolismo , Raízes de Plantas/anatomia & histologia , Raízes de Plantas/efeitos dos fármacos , Raízes de Plantas/crescimento & desenvolvimento , Estômatos de Plantas/citologia , Estômatos de Plantas/efeitos dos fármacos , Estômatos de Plantas/fisiologia , Sulfonamidas/farmacologia
15.
Neurochem Res ; 41(3): 534-43, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26198194

RESUMO

The efficient retrieval of synaptic vesicle membrane and cargo in central nerve terminals is dependent on the efficient recruitment of a series of endocytosis modes by different patterns of neuronal activity. During intense neuronal activity the dominant endocytosis mode is activity-dependent endocytosis (ADBE). Triggering of ADBE is linked to calcineurin-mediated dynamin I dephosphorylation since the same stimulation intensities trigger both. Dynamin I dephosphorylation is maximised by a simultaneous inhibition of its kinase glycogen synthase kinase 3 (GSK3) by the protein kinase Akt, however it is unknown how increased neuronal activity is transduced into Akt activation. To address this question we determined how the activity-dependent increases in intracellular free calcium ([Ca(2+)]i) control activation of Akt. This was achieved using either trains of high frequency action potentials to evoke localised [Ca(2+)]i increases at active zones, or a calcium ionophore to raise [Ca(2+)]i uniformly across the nerve terminal. Through the use of either non-specific calcium channel antagonists or intracellular calcium chelators we found that Akt phosphorylation (and subsequent GSK3 phosphorylation) was dependent on localised [Ca(2+)]i increases at the active zone. In an attempt to determine mechanism, we antagonised either phosphatidylinositol 3-kinase (PI3K) or calmodulin. Activity-dependent phosphorylation of both Akt and GSK3 was arrested on inhibition of PI3K, but not calmodulin. Thus localised calcium influx in central nerve terminals activates PI3K via an unknown calcium sensor to trigger the activity-dependent phosphorylation of Akt and GSK3.


Assuntos
Cálcio/metabolismo , Neurônios/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Terminações Pré-Sinápticas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Potenciais de Ação , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Quelantes de Cálcio/farmacologia , Ionóforos de Cálcio/farmacologia , Calmodulina/antagonistas & inibidores , Células Cultivadas , Cerebelo/citologia , Ativação Enzimática , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Masculino , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Ratos Sprague-Dawley
16.
Heart Vessels ; 31(7): 1176-84, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26498939

RESUMO

Bepridil is an effective antiarrhythmic drug on supraventricular and ventricular arrhythmias, and inhibitor of calmodulin. Recent investigations have been elucidating that bepridil exerts antiarrhythmic effects through its acute and chronic application for patients. The aim of this study was to identify the efficacy and the potential mechanism of bepridil on the inward-rectifier potassium channel in neonatal rat cardiomyocytes in acute- and long-term conditions. Bepridil inhibited inward-rectifier potassium current (I K1) as a short-term effect with IC50 of 17 µM. Bepridil also reduced I K1 of neonatal cardiomyocytes when applied for 24 h in the culture medium with IC50 of 2.7 µM. Both a calmodulin inhibitor (W-7) and an inhibitor of calmodulin-kinase II (KN93) reduced I K1 when applied for 24 h as a long-term effect in the same fashion, suggesting that the long-term application of bepridil inhibits I K1 more potently than that of the short-term application through the inhibition of calmodulin kinase II pathway in cardiomyocytes.


Assuntos
Antiarrítmicos/farmacologia , Bepridil/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Benzilaminas/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina/antagonistas & inibidores , Calmodulina/metabolismo , Células Cultivadas , Potenciais da Membrana , Miócitos Cardíacos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos Wistar , Sulfonamidas/farmacologia , Fatores de Tempo
17.
Biochem J ; 472(2): 195-204, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26399481

RESUMO

The activity of calmodulin (CaM) is modulated not only by oscillations in the cytosolic concentration of free Ca(2+), but also by its phosphorylation status. In the present study, the role of tyrosine-phosphorylated CaM [P-(Tyr)-CaM] on the regulation of the epidermal growth factor receptor (EGFR) has been examined using in vitro assay systems. We show that phosphorylation of CaM by rat liver solubilized EGFR leads to a dramatic increase in the subsequent phosphorylation of poly-L-(Glu:Tyr) (PGT) by the receptor in the presence of ligand, both in the absence and in the presence of Ca(2+). This occurred in contrast with assays where P-(Tyr)-CaM accumulation was prevented by the presence of Ca(2+), absence of a basic cofactor required for CaM phosphorylation and/or absence of CaM itself. Moreover, an antibody against CaM, which inhibits its phosphorylation, prevented the extra ligand-dependent EGFR activation. Addition of purified P-(Tyr)-CaM, phosphorylated by recombinant c-Src (cellular sarcoma kinase) and free of non-phosphorylated CaM, obtained by affinity-chromatography using an immobilized anti-phospho-(Tyr)-antibody, also increased the ligand-dependent tyrosine kinase activity of the isolated EGFR toward PGT. Also a CaM(Y99D/Y138D) mutant mimicked the effect of P-(Tyr)-CaM on ligand-dependent EGFR activation. Finally, we demonstrate that P-(Tyr)-CaM binds to the same site ((645)R-R-R-H-I-V-R-K-R-T-L-R-R-L-L-Q(660)) as non-phosphorylated CaM, located at the cytosolic juxtamembrane region of the EGFR. These results show that P-(Tyr)-CaM is an activator of the EGFR and suggest that it could contribute to the CaM-mediated ligand-dependent activation of the receptor that we previously reported in living cells.


Assuntos
Calmodulina/metabolismo , Membrana Celular/metabolismo , Receptores ErbB/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Tirosina/metabolismo , Substituição de Aminoácidos , Animais , Sítios de Ligação , Calmodulina/antagonistas & inibidores , Calmodulina/genética , Calmodulina/isolamento & purificação , Linhagem Celular Tumoral , Membrana Celular/enzimologia , Receptores ErbB/química , Receptores ErbB/genética , Receptores ErbB/isolamento & purificação , Humanos , Ligantes , Masculino , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/isolamento & purificação , Proteínas do Tecido Nervoso/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sus scrofa
18.
Exp Parasitol ; 170: 109-115, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27693219

RESUMO

FH22 has been previously identified as a calcium-binding protein from the common liver fluke, Fasciola hepatica. It is part of a family of at least four proteins in this organism which combine an EF-hand containing N-terminal domain with a C-terminal dynein light chain-like domain. Here we report further biochemical properties of FH22, which we propose should be renamed FhCaBP1 for consistency with other family members. Molecular modelling predicted that the two domains are linked by a flexible region and that the second EF-hand in the N-terminal domain is most likely the calcium ion binding site. Native gel electrophoresis demonstrated that the protein binds both calcium and manganese ions, but not cadmium, magnesium, strontium, barium, cobalt, copper(II), iron (II), nickel, zinc, lead or potassium ions. Calcium ion binding alters the conformation of the protein and increases its stability towards thermal denaturation. FhCaBP1 is a dimer in solution and calcium ions have no detectable effect on the protein's ability to dimerise. FhCaBP1 binds to the calmodulin antagonists trifluoperazine and chlorpromazine. Overall, the FhCaBP1's biochemical properties are most similar to FhCaBP2 a fact consistent with the close sequence and predicted structural similarity between the two proteins.


Assuntos
Proteínas de Ligação ao Cálcio/química , Dineínas/química , Fasciola hepatica/metabolismo , Proteínas de Helminto/química , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/isolamento & purificação , Proteínas de Ligação ao Cálcio/metabolismo , Calmodulina/antagonistas & inibidores , Clorpromazina/metabolismo , Eletroforese em Gel de Poliacrilamida , Fasciola hepatica/genética , Expressão Gênica , Proteínas de Helminto/genética , Proteínas de Helminto/isolamento & purificação , Proteínas de Helminto/metabolismo , Manganês/metabolismo , Conformação Proteica , Multimerização Proteica , Trifluoperazina/metabolismo
19.
J Psychosoc Nurs Ment Health Serv ; 54(1): 20-2, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26760133

RESUMO

Trifluoperazine, developed in the mid-1950s and introduced in 1959 as an anxiolytic and antipsychotic agent, has been an extensively studied drug molecule that has been used as a calmodulin inhibitor. Regulation of calmodulin has important roles in cellular proliferation, inflammation, neurodegeneration, and other pathological processes. Trifluoperazine also inhibits P-glycoprotein, a protein that transports organic compounds across cell membranes and the blood-brain barrier. Trifluoperazine is currently approved for the treatment of schizophrenia as well as for the treatment of non-psychotic anxiety, but has other potential clinical uses based on calmodulin and P-glycoprotein inhibition.


Assuntos
Antipsicóticos/farmacologia , Calmodulina/antagonistas & inibidores , Trifluoperazina/farmacologia , Antipsicóticos/uso terapêutico , Transtornos de Ansiedade/tratamento farmacológico , Humanos , Esquizofrenia/tratamento farmacológico , Trifluoperazina/uso terapêutico
20.
Environ Microbiol ; 17(11): 4484-94, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25970691

RESUMO

Phenylalanine ammonia lyase (PAL, E.C. 4.3.1.5) catalyses the deamination of L -phenylalanine to trans-cinnamic acid and ammonia, facilitating a critical step in the phenylpropanoid pathway that produces a variety of secondary metabolites. In this study, we isolated BbPAL gene in the entomopathogenic fungus Beauveria bassiana. According to multiple sequence alignment, homology modelling and in vitro PAL activity, we demonstrated that BbPAL acts as a typical PAL enzyme in B. bassiana. BbPAL interacted with calmodulin (CaM) in vitro and in vivo, indicating that BbPAL is a novel CaM-binding protein. The functional role of CaM in BbPAL action was to negatively regulate the BbPAL activity in B. bassiana. High-performance liquid chromatography analysis revealed that L -phenylalanine was reduced and trans-cinnamic acid was increased in response to the CaM inhibitor W-7. Dark conditions suppressed BbPAL activity in B. bassiana, compared with light. In addition, heat and cold stresses inhibited BbPAL activity in B. bassiana. Interestingly, these negative effects of BbPAL activity by dark, heat and cold conditions were recovered by W-7 treatment, suggesting that the inhibitory mechanism is mediated through stimulation of CaM activity. Therefore, this work suggests that BbPAL plays a role in the phenylpropanoid pathway mediated by environmental stimuli via the CaM signalling pathway.


Assuntos
Beauveria/metabolismo , Calmodulina/metabolismo , Insetos/microbiologia , Fenilalanina Amônia-Liase/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Calmodulina/antagonistas & inibidores , Cromatografia Líquida de Alta Pressão , Cinamatos/química , Resposta ao Choque Frio , DNA Fúngico/genética , Resposta ao Choque Térmico , Dados de Sequência Molecular , Fenilalanina/química , Fenilalanina Amônia-Liase/genética , Ligação Proteica , Alinhamento de Sequência , Transdução de Sinais/fisiologia , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA