Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Brain ; 141(7): 1998-2013, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29878067

RESUMO

Cerebellar atrophy is a key neuroradiological finding usually associated with cerebellar ataxia and cognitive development defect in children. Unlike the adult forms, early onset cerebellar atrophies are classically described as mostly autosomal recessive conditions and the exact contribution of de novo mutations to this phenotype has not been assessed. In contrast, recent studies pinpoint the high prevalence of pathogenic de novo mutations in other developmental disorders such as intellectual disability, autism spectrum disorders and epilepsy. Here, we investigated a cohort of 47 patients with early onset cerebellar atrophy and/or hypoplasia using a custom gene panel as well as whole exome sequencing. De novo mutations were identified in 35% of patients while 27% had mutations inherited in an autosomal recessive manner. Understanding if these de novo events act through a loss or a gain of function effect is critical for treatment considerations. To gain a better insight into the disease mechanisms causing these cerebellar defects, we focused on CACNA1G, a gene not yet associated with the early-onset form. This gene encodes the Cav3.1 subunit of T-type calcium channels highly expressed in Purkinje neurons and deep cerebellar nuclei. We identified four patients with de novo CACNA1G mutations. They all display severe motor and cognitive impairment, cerebellar atrophy as well as variable features such as facial dysmorphisms, digital anomalies, microcephaly and epilepsy. Three subjects share a recurrent c.2881G>A/p.Ala961Thr variant while the fourth patient has the c.4591A>G/p.Met1531Val variant. Both mutations drastically impaired channel inactivation properties with significantly slower kinetics (∼5 times) and negatively shifted potential for half-inactivation (>10 mV). In addition, these two mutations increase neuronal firing in a cerebellar nuclear neuron model and promote a larger window current fully inhibited by TTA-P2, a selective T-type channel blocker. This study highlights the prevalence of de novo mutations in early-onset cerebellar atrophy and demonstrates that A961T and M1531V are gain of function mutations. Moreover, it reveals that aberrant activity of Cav3.1 channels can markedly alter brain development and suggests that this condition could be amenable to treatment.


Assuntos
Canais de Cálcio Tipo T/genética , Ataxia Cerebelar/genética , Adolescente , Adulto , Atrofia/patologia , Encéfalo/patologia , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio Tipo T/metabolismo , Ataxia Cerebelar/fisiopatologia , Doenças Cerebelares/complicações , Cerebelo/patologia , Criança , Pré-Escolar , Estudos de Coortes , Deficiências do Desenvolvimento/genética , Feminino , Mutação com Ganho de Função/genética , Humanos , Deficiência Intelectual/genética , Masculino , Microcefalia/genética , Mutação , Linhagem , Fenótipo , Células de Purkinje/patologia
2.
J Biol Chem ; 292(49): 20010-20031, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-28972185

RESUMO

Calcium (Cav1 and Cav2) and sodium channels possess homologous CaM-binding motifs, known as IQ motifs in their C termini, which associate with calmodulin (CaM), a universal calcium sensor. Cav3 T-type channels, which serve as pacemakers of the mammalian brain and heart, lack a C-terminal IQ motif. We illustrate that T-type channels associate with CaM using co-immunoprecipitation experiments and single particle cryo-electron microscopy. We demonstrate that protostome invertebrate (LCav3) and human Cav3.1, Cav3.2, and Cav3.3 T-type channels specifically associate with CaM at helix 2 of the gating brake in the I-II linker of the channels. Isothermal titration calorimetry results revealed that the gating brake and CaM bind each other with high-nanomolar affinity. We show that the gating brake assumes a helical conformation upon binding CaM, with associated conformational changes to both CaM lobes as indicated by amide chemical shifts of the amino acids of CaM in 1H-15N HSQC NMR spectra. Intact Ca2+-binding sites on CaM and an intact gating brake sequence (first 39 amino acids of the I-II linker) were required in Cav3.2 channels to prevent the runaway gating phenotype, a hyperpolarizing shift in voltage sensitivities and faster gating kinetics. We conclude that the presence of high-nanomolar affinity binding sites for CaM at its universal gating brake and its unique form of regulation via the tuning of the voltage range of activity could influence the participation of Cav3 T-type channels in heart and brain rhythms. Our findings may have implications for arrhythmia disorders arising from mutations in the gating brake or CaM.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Calmodulina/fisiologia , Caveolina 3/metabolismo , Ativação do Canal Iônico , Animais , Sítios de Ligação , Encéfalo/fisiologia , Cálcio/metabolismo , Calmodulina/metabolismo , Coração/fisiologia , Humanos , Invertebrados , Periodicidade
3.
Proc Natl Acad Sci U S A ; 112(44): 13705-10, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26483470

RESUMO

Phosphorylation is a major mechanism regulating the activity of ion channels that remains poorly understood with respect to T-type calcium channels (Cav3). These channels are low voltage-activated calcium channels that play a key role in cellular excitability and various physiological functions. Their dysfunction has been linked to several neurological disorders, including absence epilepsy and neuropathic pain. Recent studies have revealed that T-type channels are modulated by a variety of serine/threonine protein kinase pathways, which indicates the need for a systematic analysis of T-type channel phosphorylation. Here, we immunopurified Cav3.2 channels from rat brain, and we used high-resolution MS to construct the first, to our knowledge, in vivo phosphorylation map of a voltage-gated calcium channel in a mammalian brain. We identified as many as 34 phosphorylation sites, and we show that the vast majority of these sites are also phosphorylated on the human Cav3.2 expressed in HEK293T cells. In patch-clamp studies, treatment of the channel with alkaline phosphatase as well as analysis of dephosphomimetic mutants revealed that phosphorylation regulates important functional properties of Cav3.2 channels, including voltage-dependent activation and inactivation and kinetics. We also identified that the phosphorylation of a locus situated in the loop I-II S442/S445/T446 is crucial for this regulation. Our data show that Cav3.2 channels are highly phosphorylated in the mammalian brain and establish phosphorylation as an important mechanism involved in the dynamic regulation of Cav3.2 channel gating properties.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Ativação do Canal Iônico , Canais de Cálcio Tipo T/fisiologia , Células HEK293 , Humanos , Técnicas de Patch-Clamp , Fosforilação
4.
Circulation ; 134(7): 534-46, 2016 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-27486162

RESUMO

BACKGROUND: L-type calcium channels (LTCCs) play important roles in regulating cardiomyocyte physiology, which is governed by appropriate LTCC trafficking to and density at the cell surface. Factors influencing the expression, half-life, subcellular trafficking, and gating of LTCCs are therefore critically involved in conditions of cardiac physiology and disease. METHODS: Yeast 2-hybrid screenings, biochemical and molecular evaluations, protein interaction assays, fluorescence microscopy, structural molecular modeling, and functional studies were used to investigate the molecular mechanisms through which the LTCC Cavß2 chaperone regulates channel density at the plasma membrane. RESULTS: On the basis of our previous results, we found a direct linear correlation between the total amount of the LTCC pore-forming Cavα1.2 and the Akt-dependent phosphorylation status of Cavß2 both in a mouse model of diabetic cardiac disease and in 6 diabetic and 7 nondiabetic cardiomyopathy patients with aortic stenosis undergoing aortic valve replacement. Mechanistically, we demonstrate that a conformational change in Cavß2 triggered by Akt phosphorylation increases LTCC density at the cardiac plasma membrane, and thus the inward calcium current, through a complex pathway involving reduction of Cavα1.2 retrograde trafficking and protein degradation through the prevention of dynamin-mediated LTCC endocytosis; promotion of Cavα1.2 anterograde trafficking by blocking Kir/Gem-dependent sequestration of Cavß2, thus facilitating the chaperoning of Cavα1.2; and promotion of Cavα1.2 transcription by the prevention of Kir/Gem-mediated shuttling of Cavß2 to the nucleus, where it limits the transcription of Cavα1.2 through recruitment of the heterochromatin protein 1γ epigenetic repressor to the Cacna1c promoter. On the basis of this mechanism, we developed a novel mimetic peptide that, through targeting of Cavß2, corrects LTCC life-cycle alterations, facilitating the proper function of cardiac cells. Delivery of mimetic peptide into a mouse model of diabetic cardiac disease associated with LTCC abnormalities restored impaired calcium balance and recovered cardiac function. CONCLUSIONS: We have uncovered novel mechanisms modulating LTCC trafficking and life cycle and provide proof of concept for the use of Cavß2 mimetic peptide as a novel therapeutic tool for the improvement of cardiac conditions correlated with alterations in LTCC levels and function.


Assuntos
Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/metabolismo , Canais de Cálcio Tipo L/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Peptidomiméticos/administração & dosagem , Peptidomiméticos/metabolismo , Sequência de Aminoácidos , Animais , Materiais Biomiméticos/química , Canais de Cálcio Tipo L/genética , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/metabolismo , Células Cultivadas , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Peptidomiméticos/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Estudos Retrospectivos
5.
Mol Pharmacol ; 85(2): 218-25, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24214826

RESUMO

T-type calcium channels (T/Ca(v)3-channels) are implicated in various physiologic and pathophysiologic processes such as epilepsy, sleep disorders, hypertension, and cancer. T-channels are the target of endogenous signaling lipids including the endocannabinoid anandamide, the ω3-fatty acids, and the lipoamino-acids. However, the precise molecular mechanism by which these molecules inhibit T-current is unknown. In this study, we provided a detailed electrophysiologic and pharmacologic analysis indicating that the effects of the major N-acyl derivatives on the Ca(v)3.3 current share many similarities with those of TTA-A2 [(R)-2-(4-cyclopropylphenyl)-N-(1-(5-(2,2,2-trifluoroethoxy)pyridin-2-yl)ethyl)acetamide], a synthetic T-channel inhibitor. Using radioactive binding assays with the TTA-A2 derivative [(3)H]TTA-A1 [(R)-2-(4-(tert-butyl)phenyl)-N-(1-(5-methoxypyridin-2-yl)ethyl)acetamide], we demonstrated that polyunsaturated lipids, which inhibit the Ca(v)3.3 current, as NAGly (N-arachidonoyl glycine), NASer (N-arachidonoyl-l-serine), anandamide, NADA (N-arachidonoyl dopamine), NATau (N-arachidonoyl taurine), and NA-5HT (N-arachidonoyl serotonin), all displaced [(3)H]TTA-A1 binding to membranes prepared from cells expressing Ca(v)3.3, with Ki in a micromolar or submicromolar range. In contrast, lipids with a saturated alkyl chain, as N-arachidoyl glycine and N-arachidoyl ethanolamine, which did not inhibit the Ca(v)3.3 current, had no effect on [(3)H]TTA-A1 binding. Accordingly, bio-active lipids occluded TTA-A2 effect on Ca(v)3.3 current. In addition, TTA-Q4 [(S)-4-(6-chloro-4-cyclopropyl-3-(2,2-difluoroethyl)-2-oxo-1,2,3,4-tetrahydroquinazolin-4-yl)benzonitrile], a positive allosteric modulator of [(3)H]TTA-A1 binding and TTA-A2 functional inhibition, acted in a synergistic manner to increase lipid-induced inhibition of the Ca(v)3.3 current. Overall, our results demonstrate a common molecular mechanism for the synthetic T-channel inhibitors and the endogenous lipids, and indicate that TTA-A2 and TTA-Q4 could be important pharmacologic tools to dissect the involvement of T-current in the physiologic effects of endogenous lipids.


Assuntos
Benzenoacetamidas/farmacologia , Canais de Cálcio Tipo T/fisiologia , Lipídeos/fisiologia , Piridinas/farmacologia , Regulação Alostérica , Ácidos Araquidônicos/farmacologia , Benzenoacetamidas/metabolismo , Canais de Cálcio Tipo T/efeitos dos fármacos , Células Cultivadas , Dopamina/análogos & derivados , Dopamina/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Humanos , Piridinas/metabolismo
6.
Pflugers Arch ; 466(4): 689-700, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24531745

RESUMO

T-type calcium channels (T-channels/CaV3) have unique biophysical properties allowing a calcium influx at resting membrane potential of most cells. T-channels are ubiquitously expressed in many tissues and contribute to low-threshold spikes and burst firing in central neurons as well as to pacemaker activities in cardiac cells. They also emerged as potential targets to treat cancer and hypertension. Regulation of these channels appears complex, and several studies have indicated that CaV3.1, CaV3.2, and CaV3.3 currents are directly inhibited by multiple endogenous lipids independently of membrane receptors or intracellular pathways. These bioactive lipids include arachidonic acid and ω3 poly-unsaturated fatty acids; the endocannabinoid anandamide and other N-acylethanolamides; the lipoamino-acids and lipo-neurotransmitters; the P450 epoxygenase metabolite 5,6-epoxyeicosatrienoic acid; as well as similar molecules with 18-22 carbons in the alkyl chain. In this review, we summarize evidence for direct effects of these signaling molecules, the molecular mechanisms underlying the current inhibition, and the involved chemical features. The impact of this modulation in physiology and pathophysiology is discussed with a special emphasis on pain aspects and vasodilation. Overall, these data clearly indicate that T-current inhibition is an important mechanism by which bioactive lipids mediate their physiological functions.


Assuntos
Ácido Araquidônico/fisiologia , Canais de Cálcio Tipo T/fisiologia , Endocanabinoides/fisiologia , Ácidos Graxos/fisiologia , Animais , Ácido Araquidônico/química , Endocanabinoides/química , Ácidos Graxos/química , Humanos , Lipídeos/fisiologia , Neurotransmissores/química , Neurotransmissores/fisiologia
7.
Pflugers Arch ; 466(9): 1759-68, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24327205

RESUMO

T-type calcium channels (T-channels) are important actors in neuronal pacemaking, in heart rhythm, and in the control of the vascular tone. T-channels are regulated by several endogenous lipids including the primary eicosanoid arachidonic acid (AA), which display an important role in vasodilation via its metabolism leading to prostanoids, leukotrienes, and epoxyeicosatrienoic acids (EETs). However, the effects of these latter molecules on T-currents have not been investigated. Here, we describe the effects of the major cyclooxygenase, lipoxygenase, and cytochrome P450 epoxygenase products on the three human recombinant T-channels (Cav3.1, Cav3.2, and Cav3.3), as compared to those of AA. We identified the P450 epoxygenase product, 5,6-EET, as a potent physiological inhibitor of Cav3 currents. The effects of 5,6-EET were observed at sub-micromolar concentrations (IC50 = 0.54 µM), occurred in the minute range, and were reversible. The 5,6-EET inhibited the Cav3 currents at physiological resting membrane potentials mostly by inducing a large negative shift in their steady-state inactivation properties. Using knockout mice for Cav3.1 and Cav3.2, we demonstrated that the vasodilation of preconstricted mesenteric arteries induced by 5,6-EET was specifically impaired in Cav3.2 knockout mice. Overall, our results indicate that inhibition of Cav3 currents by 5,6-EET is an important mechanism controlling the vascular tone.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Canais de Cálcio Tipo T/metabolismo , Músculo Liso Vascular/metabolismo , Ácido 8,11,14-Eicosatrienoico/metabolismo , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Canais de Cálcio Tipo T/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tono Muscular/efeitos dos fármacos , Tono Muscular/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Técnicas de Patch-Clamp , Transfecção
8.
iScience ; 27(4): 109396, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38510134

RESUMO

The recent development of single-cell and single-nucleus RNA sequencing has highlighted the extraordinary diversity of dorsal root ganglia neurons. However, the few available genetic tools limit our understanding of the functional significance of this heterogeneity. We generated a new mouse line expressing the flippase recombinase from the scn10a locus. By crossing Nav1.8Ires-FLPo mice with the AdvillinCre and RC::FL-hM3Dq mouse lines in an intersectional genetics approach, we were able to obtain somatodendritic expression of hM3Dq-mCherry selectively in the Nav1.8 lineage. The bath application of clozapine N-oxide triggered strong calcium responses selectively in mCherry+ neurons. The intraplantar injection of CNO caused robust flinching, shaking, and biting responses accompanied by strong cFos activation in the ipsilateral lumbar spinal cord. The Nav1.8Ires-FLPo mouse model will be a valuable tool for extending our understanding of the in vivo functional specialization of neuronal subsets of the Nav1.8 lineage for which inducible Cre lines are available.

9.
RSC Adv ; 14(3): 1750-1756, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38192327

RESUMO

Herein, we demonstrate the use of an atmospheric pressure plasma with a Dielectric Barrier Discharge (DBD) for the synthesis of FeOx nanoparticles with a simultaneous formation of graphene oxide domains at low substrate temperature. For that, the interaction of the plasma to control good decomposition of the Fe precursor is essential and this is demonstrated by FTIR analyses. Thanks to a fine tuning of the plasma conditions, a homogeneous spatial distribution around 5 nm nanoparticles (NPs) was obtained, whereas without plasma, in the same configuration of the process, a heterogeneity regarding size and shape for the NPs was obtained. The Raman spectrum of the plasma deposit confirmed the presence of graphene oxide as the characteristic G and D bands were observed with I(D)/I(G) = 0.92. Thanks to optical emission spectroscopy (OES) measurements, it is proposed that the carbon deposition on FeOx nanoparticles is produced on the near plasma post discharge. XPS studies showed that the main contribution of iron was in Fe2+ form, corresponding to the FeO phase. No metallic Fe or carbide were detected. As there are many studies reporting the synergetic effect of FeOx NPs and graphene oxide, we believe that this new one-step simultaneous synthesis method may be of high interest for applications requiring direct deposition on temperature labile substrates such as polymers.

10.
Br J Pharmacol ; 181(12): 1793-1811, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38369690

RESUMO

BACKGROUND AND PURPOSE: Voltage sensitivity is a common feature of many membrane proteins, including some G-protein coupled receptors (GPCRs). However, the functional consequences of voltage sensitivity in GPCRs are not well understood. EXPERIMENTAL APPROACH: In this study, we investigated the voltage sensitivity of the post-synaptic metabotropic glutamate receptor mGlu5 and its impact on synaptic transmission. Using biosensors and electrophysiological recordings in non-excitable HEK293T cells or neurons. KEY RESULTS: We found that mGlu5 receptor function is optimal at resting membrane potentials. We observed that membrane depolarization significantly reduced mGlu5 receptor activation, Gq-PLC/PKC stimulation, Ca2+ release and mGlu5 receptor-gated currents through transient receptor potential canonical, TRPC6, channels or glutamate ionotropic NMDA receptors. Notably, we report a previously unknown activity of the NMDA receptor at the resting potential of neurons, enabled by mGlu5. CONCLUSIONS AND IMPLICATIONS: Our findings suggest that mGlu5 receptor activity is directly regulated by membrane voltage which may have a significant impact on synaptic processes and pathophysiological functions.


Assuntos
Receptor de Glutamato Metabotrópico 5 , Transmissão Sináptica , Animais , Humanos , Células HEK293 , Potenciais da Membrana , Neurônios/metabolismo , Neurônios/fisiologia , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia , Camundongos
11.
Nat Commun ; 15(1): 54, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-38167790

RESUMO

L-type voltage-gated calcium channels are involved in multiple physiological functions. Currently available antagonists do not discriminate between L-type channel isoforms. Importantly, no selective blocker is available to dissect the role of L-type isoforms Cav1.2 and Cav1.3 that are concomitantly co-expressed in the heart, neuroendocrine and neuronal cells. Here we show that calciseptine, a snake toxin purified from mamba venom, selectively blocks Cav1.2 -mediated L-type calcium currents (ICaL) at concentrations leaving Cav1.3-mediated ICaL unaffected in both native cardiac myocytes and HEK-293T cells expressing recombinant Cav1.2 and Cav1.3 channels. Functionally, calciseptine potently inhibits cardiac contraction without altering the pacemaker activity in sino-atrial node cells, underscoring differential roles of Cav1.2- and Cav1.3 in cardiac contractility and automaticity. In summary, calciseptine is a selective L-type Cav1.2 Ca2+ channel blocker and should be a valuable tool to dissect the role of these L-channel isoforms.


Assuntos
Canais de Cálcio Tipo L , Dendroaspis , Animais , Canais de Cálcio Tipo L/fisiologia , Dendroaspis/metabolismo , Miócitos Cardíacos/metabolismo , Isoformas de Proteínas , Cálcio/metabolismo
12.
J Neurosci ; 32(12): 4284-96, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22442090

RESUMO

Ligand-gated ion channels are prototypic oligomeric membrane proteins whose stoichiometry determines their functional properties and subcellular localization. Deciphering the quaternary structure of such protein complexes is an arduous task and usually requires the combination of multiple approaches. ATP-gated P2X receptors are formed by the association of three subunits, but the quaternary arrangement of the seven P2X subunits at the plasma membrane remains poorly characterized. By combining bioluminescence resonance energy transfer, bifunctional fluorescence complementation and protein biochemistry, we developed an experimental approach that allows precise determination of rat P2X receptor quaternary assembly. We found that P2X5 subunits associate with P2X1, P2X2, and P2X4 subunits. We demonstrate that P2X5 and P2X2 subunits interact to form as yet uncharacterized heteromeric receptors with alternate stoichiometries, both present at the plasma membrane. P2X2/5 receptors display functional properties such as pore dilatation, membrane blebbing, and phosphatidylserine exposure that were previously thought to be characteristic hallmarks of the P2X7 receptor. In mouse, P2X2 and P2X5 subunits colocalize and physically interact in specific neuronal populations suggesting that other P2X receptors might contribute to cellular responses typically attributed to P2X7 receptor.


Assuntos
Subunidades Proteicas/metabolismo , Receptores Purinérgicos P2X2/metabolismo , Receptores Purinérgicos P2X5/metabolismo , Receptores Purinérgicos P2X7/fisiologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Anexina A5/metabolismo , Benzoxazóis/metabolismo , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Encéfalo/metabolismo , Ensaio de Imunoadsorção Enzimática , Gânglios Espinais/citologia , Células HEK293 , Humanos , Imunoprecipitação , Proteínas Luminescentes/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Mutagênese Sítio-Dirigida/métodos , Mutação/genética , Técnicas de Patch-Clamp , Subunidades Proteicas/genética , Purinérgicos/farmacologia , Compostos de Quinolínio/metabolismo , Receptores Purinérgicos P2X2/genética , Receptores Purinérgicos P2X5/genética , Transfecção , Gravação em Vídeo , Xenopus laevis
13.
Nanoscale Adv ; 5(9): 2573-2582, 2023 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-37143807

RESUMO

Herein, we report the impact of plasma on gold nanoparticles synthesis. We used an atmospheric plasma torch fed with an aerosolized tetrachloroauric(iii) acid trihydrate (HAuCl4·3H2O) solution. The investigation showed that using pure ethanol as a solvent for the gold precursor enabled a better dispersion compared to a water-containing solution. We demonstrated here that the deposition parameters are easy to control, presenting the influence of solvent concentration and deposition time. The advantage of our method is that no capping agent was used. We assume that plasma creates a carbon-based matrix around the gold nanoparticles preventing them to agglomerate. The XPS results revealed the impact of using plasma. Metallic gold was detected in the plasma-treated sample, whereas the no-plasma sample revealed only Au(i) and Au(iii) contributions originating from the HAuCl4 precursor. Detailed HRTEM, EDS mapping, and SAED analyses led to more insights into the structure.

14.
ACS Omega ; 8(6): 5475-5485, 2023 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-36816692

RESUMO

Gallium-doped zinc oxide (GZO) films were fabricated using RF magnetron sputtering and atomic layer deposition (ALD). The latter ones demonstrate higher electrical conductivities (up to 2700 S cm-1) and enhanced charge mobilities (18 cm2 V-1 s-1). The morphological analysis reveals differences mostly due to the very different nature of the deposition processes. The film deposited via ALD shows an increased transmittance in the visible range and a very small one in the infrared range that leads to a figure of merit of 0.009 Ω-1 (10 times higher than for the films deposited via sputtering). A benchmarking is made with an RF sputtered indium-doped tin oxide (ITO) film used conventionally in the industry. Another comparison between ZnO, Al:ZnO (AZO), and Ga:ZnO (GZO) films fabricated by ALD is presented, and the evolution of physical properties with doping is evidenced. Finally, we processed GZO thin films on a glass substrate into patterned transparent patch antennas to demonstrate an application case of short-range communication by means of the Bluetooth Low Energy (BLE) protocol. The GZO transparent antennas' performances are compared to a reference ITO antenna on a glass substrate and a conventional copper antenna on FR4 PCB. The results highlight the possibility to use the transparent GZO antenna for reliable short-range communication and the achievability of an antenna entirely processed by ALD.

15.
FEBS J ; 290(14): 3688-3702, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36912793

RESUMO

Venom-derived peptides targeting ion channels involved in pain are regarded as a promising alternative to current, and often ineffective, chronic pain treatments. Many peptide toxins are known to specifically and potently block established therapeutic targets, among which the voltage-gated sodium and calcium channels are major contributors. Here, we report on the discovery and characterization of a novel spider toxin isolated from the crude venom of Pterinochilus murinus that shows inhibitory activity at both hNaV 1.7 and hCaV 3.2 channels, two therapeutic targets implicated in pain pathways. Bioassay-guided HPLC fractionation revealed a 36-amino acid peptide with three disulfide bridges named µ/ω-theraphotoxin-Pmu1a (Pmu1a). Following isolation and characterization, the toxin was chemically synthesized and its biological activity was further assessed using electrophysiology, revealing Pmu1a to be a toxin that potently blocks both hNaV 1.7 and hCaV 3. Nuclear magnetic resonance structure determination of Pmu1a shows an inhibitor cystine knot fold that is the characteristic of many spider peptides. Combined, these data show the potential of Pmu1a as a basis for the design of compounds with dual activity at the therapeutically relevant hCaV 3.2 and hNaV 1.7 voltage-gated channels.


Assuntos
Venenos de Aranha , Aranhas , Animais , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Venenos de Aranha/farmacologia , Venenos de Aranha/química , Venenos de Aranha/metabolismo , Dor , Peptídeos/farmacologia , Espectroscopia de Ressonância Magnética , Aranhas/metabolismo
16.
Sci Adv ; 8(26): eabo7566, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35767616

RESUMO

Affective touch is necessary for proper neurodevelopment and sociability. However, it remains unclear how the neurons innervating the skin detect affective and social behaviors. The C low-threshold mechanoreceptors (C-LTMRs), a specific population of somatosensory neurons in mice, appear particularly well suited, physiologically and anatomically, to perceive affective and social touch. However, their contribution to sociability has not been resolved yet. Our observations revealed that C-LTMR functional deficiency induced social isolation and reduced tactile interactions in adulthood. Conversely, transient increase in C-LTMR excitability in adults, using chemogenetics, was rewarding, promoted touch-seeking behaviors, and had prosocial influences on group dynamics. This work provides the first empirical evidence that specific peripheral inputs alone can drive complex social behaviors. It demonstrates the existence of a specialized neuronal circuit, originating in the skin, wired to promote interactions with other individuals.

17.
EMBO Rep ; 10(8): 873-80, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19575010

RESUMO

A previously uncharacterized putative ion channel, NALCN (sodium leak channel, non-selective), has been recently shown to be responsible for the tetrodotoxin (TTX)-resistant sodium leak current implicated in the regulation of neuronal excitability. Here, we show that NALCN encodes a current that is activated by M3 muscarinic receptors (M3R) in a pancreatic beta-cell line. This current is primarily permeant to sodium ions, independent of intracellular calcium stores and G proteins but dependent on Src activation, and resistant to TTX. The current is recapitulated by co-expression of NALCN and M3R in human embryonic kidney-293 cells and in Xenopus oocytes. We also show that NALCN and M3R belong to the same protein complex, involving the intracellular I-II loop of NALCN and the intracellular i3 loop of M3R. Taken together, our data show the molecular basis of a muscarinic-activated inward sodium current that is independent of G-protein activation, and provide new insights into the properties of NALCN channels.


Assuntos
Células Secretoras de Insulina/metabolismo , Canais Iônicos/metabolismo , Receptor Muscarínico M3/metabolismo , Western Blotting , Linhagem Celular , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Canais Iônicos/genética , Técnicas de Patch-Clamp , Pirazóis/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Receptor Muscarínico M3/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Neurosci ; 29(42): 13106-14, 2009 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-19846698

RESUMO

Lipoamino acids are anandamide-related endogenous molecules that induce analgesia via unresolved mechanisms. Here, we provide evidence that the T-type/Cav3 calcium channels are important pharmacological targets underlying their physiological effects. Various lipoamino acids, including N-arachidonoyl glycine (NAGly), reversibly inhibited Cav3.1, Cav3.2, and Cav3.3 currents, with potent effects on Cav3.2 [EC(50) approximately 200 nm for N-arachidonoyl 3-OH-gamma-aminobutyric acid (NAGABA-OH)]. This inhibition involved a large shift in the Cav3.2 steady-state inactivation and persisted during fatty acid amide hydrolase (FAAH) inhibition as well as in cell-free outside-out patch. In contrast, lipoamino acids had weak effects on high-voltage-activated (HVA) Cav1.2 and Cav2.2 calcium currents, on Nav1.7 and Nav1.8 sodium currents, and on anandamide-sensitive TRPV1 and TASK1 currents. Accordingly, lipoamino acids strongly inhibited native Cav3.2 currents in sensory neurons with small effects on sodium and HVA calcium currents. In addition, we demonstrate here that lipoamino acids NAGly and NAGABA-OH produced a strong thermal analgesia and that these effects (but not those of morphine) were abolished in Cav3.2 knock-out mice. Collectively, our data revealed lipoamino acids as a family of endogenous T-type channel inhibitors, suggesting that these ligands can modulate multiple cell functions via this newly evidenced regulation.


Assuntos
Analgésicos/farmacologia , Ácidos Araquidônicos/farmacologia , Canais de Cálcio Tipo T/metabolismo , Glicina/análogos & derivados , Ácido gama-Aminobutírico/análogos & derivados , Animais , Comportamento Animal/fisiologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo T/classificação , Canais de Cálcio Tipo T/genética , Células Cultivadas , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Gânglios Espinais/citologia , Glicina/farmacologia , Proteínas de Fluorescência Verde/genética , Humanos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/genética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7 , Proteínas do Tecido Nervoso/genética , Neuroblastoma , Técnicas de Patch-Clamp/métodos , Canais de Potássio de Domínios Poros em Tandem/genética , Células Receptoras Sensoriais , Canais de Sódio/genética , Canais de Cátion TRPV/genética , Transfecção , Ácido gama-Aminobutírico/farmacologia
19.
Biochim Biophys Acta ; 1793(6): 947-52, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19071165

RESUMO

T-type calcium channels (T-channels) contribute to a wide variety of physiological functions, especially in the cardiovascular and nervous systems. Recent studies using knock-out mouse models have been instrumental in documenting further the role of T-channels in sleep, heartbeat, pain and epilepsy. Importantly, several novel aspects of the regulation of these channels have been identified over the last few years, providing new insights into their physiological and pathophysiological roles. Here, we review recent evidence supporting that the Cav3 subunits of T-channels are modulated by endogenous ligands such as anandamide, zinc, redox and oxidizing agents, as well as G-protein and protein kinases pathways. The study of T-channel mutations associated with childhood absence epilepsy has also revealed new aspects of Cav3 subunit trafficking. Collectively, these findings identify novel regulatory mechanisms involved in the fine tuning of T-channel expression and activity, and offer new directions for the design of novel therapeutic strategies targeting these channels.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Isoformas de Proteínas/metabolismo , Transdução de Sinais/fisiologia , Animais , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio Tipo T/química , Canais de Cálcio Tipo T/genética , Canalopatias/genética , Canalopatias/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Isoformas de Proteínas/genética , Estrutura Secundária de Proteína
20.
Sci Rep ; 9(1): 15642, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31666636

RESUMO

Cav3 / T-type Ca2+ channels are dynamically regulated by intracellular Ca2+ ions, which inhibit Cav3 availability. Here, we demonstrate that this inhibition becomes irreversible in the presence of non-hydrolysable ATP analogs, resulting in a strong hyperpolarizing shift in the steady-state inactivation of the residual Cav3 current. Importantly, the effect of these ATP analogs was prevented in the presence of intracellular BAPTA. Additional findings obtained using intracellular dialysis of inorganic phosphate and alkaline phosphatase or NaN3 treatment further support the involvement of a phosphorylation mechanism. Contrasting with Cav1 and Cav2 Ca2+ channels, the Ca2+-dependent modulation of Cav3 channels appears to be independent of calmodulin, calcineurin and endocytic pathways. Similar findings were obtained for the native T-type Ca2+ current recorded in rat thalamic neurons of the central medial nucleus. Overall, our data reveal a new Ca2+ sensitive phosphorylation-dependent mechanism regulating Cav3 channels, with potentially important physiological implications for the multiple cell functions controlled by T-type Ca2+ channels.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Cálcio/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Canais de Cálcio Tipo T/genética , Feminino , Masculino , Fosfatos/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Núcleos Talâmicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA