Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 545
Filtrar
1.
Acta Physiol (Oxf) ; 240(2): e14075, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38071417

RESUMEN

AIM: Bradyarrhythmias result from inhibition of automaticity, prolonged repolarization, or slow conduction in the heart. The ERG channels mediate the repolarizing current IKr in the cardiac action potential, whereas T-type calcium channels (TTCC) are involved in the sinoatrial pacemaker and atrioventricular conduction in mammals. Zebrafish have become a valuable research model for human cardiac electrophysiology and disease. Here, we investigate the contribution of ERG channels and TTCCs to the pacemaker and atrioventricular conduction in zebrafish larvae and determine the mechanisms causing atrioventricular block. METHODS: Zebrafish larvae expressing ratiometric fluorescent Ca2+ biosensors in the heart were used to measure Ca2+ levels and rhythm in beating hearts in vivo, concurrently with contraction and hemodynamics. The atrioventricular delay (the time between the start of atrial and ventricular Ca2+ transients) was used to measure impulse conduction velocity and distinguished between slow conduction and prolonged refractoriness as the cause of the conduction block. RESULTS: ERG blockers caused bradycardia and atrioventricular block by prolonging the refractory period in the atrioventricular canal and in working ventricular myocytes. In contrast, inhibition of TTCCs caused bradycardia and second-degree block (Mobitz type I) by slowing atrioventricular conduction. TTCC block did not affect ventricular contractility, despite being highly expressed in cardiomyocytes. Concomitant measurement of Ca2+ levels and ventricular size showed mechano-mechanical coupling: increased preload resulted in a stronger heart contraction in vivo. CONCLUSION: ERG channels and TTCCs influence the heart rate and atrioventricular conduction in zebrafish larvae. The zebrafish lines expressing Ca2+ biosensors in the heart allow us to investigate physiological feedback mechanisms and complex arrhythmias.


Asunto(s)
Bloqueo Atrioventricular , Canales de Calcio Tipo T , Marcapaso Artificial , Humanos , Animales , Pez Cebra , Frecuencia Cardíaca/fisiología , Bradicardia , Canales de Calcio Tipo T/fisiología , Canales de Potasio Éter-A-Go-Go , Miocitos Cardíacos , Mamíferos , Regulador Transcripcional ERG
2.
Nihon Yakurigaku Zasshi ; 158(3): 218-222, 2023 May 01.
Artículo en Japonés | MEDLINE | ID: mdl-36990796

RESUMEN

Alzheimer's disease (AD) is the most common dementia in the world characterized by the neuropathological hallmarks consisting of an accumulation of extracellular amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles (NFT). There is no fundamental therapeutic treatment. We have developed a novel AD therapeutic candidate SAK3 which improves neuronal plasticity in the brain. SAK3 enhanced the acetylcholine release via T-type calcium channels. T-type calcium channels is highly expressed in neuro-progenitor cells in the hippocampal dentate gyrus. SAK3 enhanced the proliferation and differentiation of the neuro-progenitor cells, thereby improving depressive behaviors. The Cav3.1 null mice impaired the proliferation and differentiation of the neuro-progenitor cells. In addition, SAK3 activated CaMKII involving neuronal plasticity, thereby improving spine regeneration and proteasome activities impaired in AD related AppNL-F/NL-F knock-in mice. The improvement of the decreased proteasome activity through enhancement CaMKII/Rpt6 signaling by SAK3 treatment contributed to the amelioration of synaptic abnormalities and cognitive decline. The increased proteasome activity also accounted for inhibition of Aß deposition. Taken together, the proteasome activation via enhancement of CaMKII/Rpt6 signaling is a new strategy for AD treatment, which rescues the AD pathology including cognitive impairments and Aß deposition. SAK3 may be a new hopeful drug candidate rescuing dementia patients.


Asunto(s)
Enfermedad de Alzheimer , Canales de Calcio Tipo T , Ratones , Animales , Canales de Calcio Tipo T/fisiología , Complejo de la Endopetidasa Proteasomal/uso terapéutico , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/uso terapéutico , Ratones Noqueados , Plasticidad Neuronal
3.
Redox Biol ; 59: 102579, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36563535

RESUMEN

Poly-trans-[(2-carboxyethyl)germasesquioxane] (Ge-132), an organogermanium, is hydrolyzed to 3-(trihydroxygermyl)propanoic acid (THGP) in aqueous solutions, and reduces inflammation, pain and cancer, whereas the underlying mechanisms remain unknown. Sulfides including H2S, a gasotransmitter, generated from l-cysteine by some enzymes including cystathionine-γ-lyase (CSE), are pro-nociceptive, since they enhance Cav3.2 T-type Ca2+ channel activity expressed in the primary afferents, most probably by canceling the channel inhibition by Zn2+ linked via coordinate bonding to His191 of Cav3.2. Given that germanium is reactive to sulfur, we tested whether THGP would directly trap sulfide, and inhibit sulfide-induced enhancement of Cav3.2 activity and sulfide-dependent pain in mice. Using mass spectrometry and 1H NMR techniques, we demonstrated that THGP directly reacted with sulfides including Na2S and NaSH, and formed a sulfur-containing reaction product, which decreased in the presence of ZnCl2. In Cav3.2-transfected HEK293 cells, THGP inhibited the sulfide-induced enhancement of T-type Ca2+ channel-dependent membrane currents. In mice, THGP, administered systemically or locally, inhibited the mechanical allodynia caused by intraplantar Na2S. In the mice with cyclophosphamide-induced cystitis and cerulein-induced pancreatitis, which exhibited upregulation of CSE in the bladder and pancreas, respectively, systemic administration of THGP as well as a selective T-type Ca2+ channel inhibitor suppressed the cystitis-related and pancreatitis-related visceral pain. These data suggest that THGP traps sulfide and inhibits sulfide-induced enhancement of Cav3.2 activity, leading to suppression of Cav3.2-dependent pain caused by sulfide applied exogenously and generated endogenously.


Asunto(s)
Canales de Calcio Tipo T , Cistitis , Sulfuro de Hidrógeno , Pancreatitis , Dolor Visceral , Ratones , Humanos , Animales , Células HEK293 , Canales de Calcio Tipo T/fisiología , Sulfuros/farmacología , Cistitis/inducido químicamente , Sulfuro de Hidrógeno/metabolismo
4.
Br J Pharmacol ; 179(15): 4031-4043, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35342937

RESUMEN

BACKGROUND AND PURPOSE: T-type Ca channels (ICa ) regulate neuronal excitability and contribute to neurotransmitter release. The phytocannabinoids Δ9 -tetrahydrocannabinol and cannabidiol effectively modulate T-type ICa , but effects of other biologically active phytocannabinoids on these channels are unknown. We thus investigated the modulation of T-type ICa by low abundance phytocannabinoids. EXPERIMENTAL APPROACH: A fluorometric (fluorescence imaging plate reader [FLIPR]) assay was used to investigate modulation of human T-type ICa (CaV 3.1, 3.2 and 3.3) stably expressed in FlpIn-TREx HEK293 cells. The biophysical effects of some compounds were examined using whole-cell patch clamp recordings. KEY RESULTS: In the FLIPR assay, all 11 phytocannabinoids tested modulated T-type ICa , with most inhibiting CaV 3.1 and CaV 3.2 more effectively than CaV 3.3. Cannabigerolic acid was the most potent inhibitor of CaV 3.1 (pIC50 6.1 ± 0.6) and CaV 3.2 (pIC50 6.4 ± 0.4); in all cases, phytocannabinoid acids were more potent than their corresponding neutral forms. In patch clamp recordings, cannabigerolic acid inhibited CaV 3.1 and 3.2 with similar potency to the FLIPR assay; the inhibition was associated with significant hyperpolarizing shift in activation and steady-state inactivation of these channels. In contrast, cannabidiol, cannabidivarin, and cannabigerol only affected channel inactivation. CONCLUSION AND IMPLICATIONS: Modulation of T-type calcium channels is a common property of phytocannabinoids, which all increase steady-state inactivation at physiological membrane potentials, with some also affecting channel activation. Thus, T-type ICa may be a common site of action for phytocannabinoids, and the diverse actions of phytocannabinoids on channel gating may provide insight into structural requirement for selective T-type ICa modulators.


Asunto(s)
Canales de Calcio Tipo T , Cannabidiol , Calcio , Canales de Calcio Tipo T/fisiología , Cannabidiol/farmacología , Células HEK293 , Humanos , Potenciales de la Membrana , Técnicas de Placa-Clamp
5.
Toxins (Basel) ; 14(2)2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35202127

RESUMEN

Conotoxins are tools used by marine Conus snails to hunt and are a significant repository for marine drug research. Conotoxins highly selectively coordinate different subtypes of various ion channels, and a few have been used in pain management. Although more than 8000 conotoxin genes have been found, the biological activity and function of most have not yet been examined. In this report, we selected the toxin gene QcMNCL-XIII0.1 from our previous investigation and studied it in vitro. First, we successfully prepared active recombinant QcMNCL-XIII0.1 using a TrxA (Thioredoxin A)-assisted folding expression vector based on genetic engineering technology. Animal experiments showed that the recombinant QcMNCL-XIII0.1 exhibited nerve conduction inhibition similar to that of pethidine hydrochloride. With flow cytometry combined fluorescent probe Fluo-4 AM, we found that 10 ng/µL recombinant QcMNCL-XIII0.1 inhibited the fluorescence intensity by 31.07% in the 293T cell model transfected with Cav3.1, implying an interaction between α1G T-type calcium channel protein and recombinant QcMNCL-XIII0.1. This toxin could be an important drug in biomedical research and medicine for pain control.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Conotoxinas/toxicidad , Conducción Nerviosa/efectos de los fármacos , Nervio Ciático/efectos de los fármacos , Animales , Canales de Calcio Tipo T/genética , Línea Celular , Conotoxinas/genética , Caracol Conus , Estimulación Eléctrica , Humanos , Rana catesbeiana , Proteínas Recombinantes/toxicidad , Nervio Ciático/fisiología
6.
Theranostics ; 11(19): 9342-9357, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34646374

RESUMEN

Background: Neuromedin B (Nmb) is implicated in the regulation of nociception of sensory neurons. However, the underlying cellular and molecular mechanisms remain unknown. Methods: Using patch clamp recording, western blot analysis, immunofluorescent labelling, enzyme-linked immunosorbent assays, adenovirus-mediated shRNA knockdown and animal behaviour tests, we studied the effects of Nmb on the sensory neuronal excitability and peripheral pain sensitivity mediated by Cav3.2 T-type channels. Results: Nmb reversibly and concentration-dependently increased T-type channel currents (IT) in small-sized trigeminal ganglion (TG) neurons through the activation of neuromedin B receptor (NmbR). This NmbR-mediated IT response was Gq protein-coupled, but independent of protein kinase C activity. Either intracellular application of the QEHA peptide or shRNA-mediated knockdown of Gß abolished the NmbR-induced IT response. Inhibition of protein kinase A (PKA) or AMP-activated protein kinase (AMPK) completely abolished the Nmb-induced IT response. Analysis of phospho-AMPK (p-AMPK) revealed that Nmb significantly activated AMPK, while AMPK inhibition prevented the Nmb-induced increase in PKA activity. In a heterologous expression system, activation of NmbR significantly enhanced the Cav3.2 channel currents, while the Cav3.1 and Cav3.3 channel currents remained unaffected. Nmb induced TG neuronal hyperexcitability and concomitantly induced mechanical and thermal hypersensitivity, both of which were attenuated by T-type channel blockade. Moreover, blockade of NmbR signalling prevented mechanical hypersensitivity in a mouse model of complete Freund's adjuvant-induced inflammatory pain, and this effect was attenuated by siRNA knockdown of Cav3.2. Conclusions: Our study reveals a novel mechanism by which NmbR stimulates Cav3.2 channels through a Gßγ-dependent AMPK/PKA pathway. In mouse models, this mechanism appears to drive the hyperexcitability of TG neurons and induce pain hypersensitivity.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Dolor/metabolismo , Receptores de Bombesina/metabolismo , Potenciales de Acción , Animales , Canales de Calcio Tipo T/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Adyuvante de Freund/farmacología , Ganglios Espinales/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Neuroquinina B/análogos & derivados , Neuroquinina B/metabolismo , Dolor/fisiopatología , Receptores de Bombesina/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Transducción de Señal/efectos de los fármacos , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo
7.
Mol Brain ; 14(1): 145, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34544471

RESUMEN

The CACNA1H gene encodes the α1 subunit of the low voltage-activated Cav3.2 T-type calcium channel, an important regulator of neuronal excitability. Alternative mRNA splicing can generate multiple channel variants with distinct biophysical properties and expression patterns. Two major splice variants, containing or lacking exon 26 (± 26) have been found in different human tissues. In this study, we report splice variant specific effects of a Cav3.2 mutation found in patients with autosomal dominant writer's cramp, a specific type of focal dystonia. We had previously reported that the R481C missense mutation caused a gain of function effect when expressed in Cav3.2 (+ 26) by accelerating its recovery from inactivation. Here, we show that when the mutation is expressed in the short variant of the channel (- 26), we observe a significant increase in current density when compared to wild-type Cav3.2 (- 26) but the effect on the recovery from inactivation is lost. Our data add to growing evidence that the functional expression of calcium channel mutations depends on which splice variant is being examined.


Asunto(s)
Canales de Calcio Tipo T/genética , Trastornos Distónicos/genética , Potenciales de Acción , Empalme Alternativo , Bario/metabolismo , Canales de Calcio Tipo T/fisiología , Trastornos Distónicos/fisiopatología , Exones/genética , Mutación con Ganancia de Función , Humanos , Transporte Iónico , Modelos Moleculares , Mutación Missense , Mutación Puntual , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología
8.
Mol Brain ; 14(1): 126, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34399820

RESUMEN

Developmental and epileptic encephalopathies (DEEs) are a group of severe epilepsies that are characterized by seizures and developmental delay. DEEs are primarily attributed to genetic causes and an increasing number of cases have been correlated with variants in ion channel genes. In this study, we report a child with an early severe DEE. Whole exome sequencing showed a de novo heterozygous variant (c.4873-4881 duplication) in the SCN8A gene and an inherited heterozygous variant (c.952G > A) in the CACNA1H gene encoding for Nav1.6 voltage-gated sodium and Cav3.2 voltage-gated calcium channels, respectively. In vitro functional analysis of human Nav1.6 and Cav3.2 channel variants revealed mild but significant alterations of their gating properties that were in general consistent with a gain- and loss-of-channel function, respectively. Although additional studies will be required to confirm the actual pathogenic involvement of SCN8A and CACNA1H, these findings add to the notion that rare ion channel variants may contribute to the etiology of DEEs.


Asunto(s)
Discapacidades del Desarrollo/genética , Epilepsia Refractaria/genética , Epilepsia Tónico-Clónica/genética , Canal de Sodio Activado por Voltaje NAV1.6/genética , Anomalías Múltiples/genética , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/fisiología , Femenino , Mutación con Ganancia de Función , Duplicación de Gen , Predisposición Genética a la Enfermedad , Humanos , Recién Nacido , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.6/fisiología , Linaje , Mutación Puntual , Escoliosis/genética
9.
Transl Res ; 234: 20-30, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33422652

RESUMEN

T-type calcium channels regulate neuronal excitability and are important contributors of pain processing. CaV3.2 channels are the major isoform expressed in nonpeptidergic and peptidergic nociceptive neurons and are emerging as promising targets for pain treatment. Numerous studies have shown that CaV3.2 expression and/or activity are significantly increased in spinal dorsal horn and in dorsal root ganglia neurons in different inflammatory and neuropathic pain models. Pharmacological campaigns to inhibit the functional expression of CaV3.2 for treatment of pain have focused on the development of direct channel blockers, but none have produced lead candidates. Targeting the proteins that regulate the trafficking or transcription, and the ones that modify the channels via post-translational modifications are alternative means to regulate expression and function of CaV3.2 channels and hence to develop new drugs to control pain. Here we synthesize data supporting a role for CaV3.2 in numerous pain modalities and then discuss emerging opportunities for the indirect targeting of CaV3.2 channels.


Asunto(s)
Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo T/fisiología , Dolor Crónico/tratamiento farmacológico , Animales , Fenómenos Biofísicos , Canales de Calcio Tipo T/química , Canales de Calcio Tipo T/genética , Dolor Crónico/fisiopatología , Modelos Animales de Enfermedad , Desarrollo de Medicamentos , Ganglios Espinales/fisiopatología , Humanos , Modelos Moleculares , Neuralgia/tratamiento farmacológico , Neuralgia/fisiopatología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Asta Dorsal de la Médula Espinal/fisiopatología , Transcripción Genética/efectos de los fármacos , Investigación Biomédica Traslacional
10.
Sci Rep ; 10(1): 15993, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33009476

RESUMEN

The mating of 77 heterozygous pairs (Cav3.2[+|-] x Cav3.2[+|-]) revealed a significant deviation of genotype distribution from Mendelian inheritance in weaned pups. The mating of 14 pairs (Cav3.2[-|-] female x Cav3.2[+|-] male) and 8 pairs (Cav3.2[+|-] female x Cav3.2[-|-] male) confirmed the significant reduction of deficient homozygous Cav3.2[-|-] pups, leading to the conclusion that prenatal lethality may occur, when one or both alleles, encoding the Cav3.2T-type Ca2+ channel, are missing. Also, the mating of 63 heterozygous pairs (Cav2.3[+|-] x Cav2.3[+|-]) revealed a significant deviation of genotype distribution from Mendelian inheritance in weaned pups, but only for heterozygous male mice, leading to the conclusion that compensation may only occur for Cav2.3[-|-] male mice lacking both alleles of the R-type Ca2+ channel. During the mating of heterozygous parents, the number of female mice within the weaned population does not deviate from the expected Mendelian inheritance. During prenatal development, both, T- and R-type Ca2+ currents are higher expressed in some tissues than postnatally. It will be discussed that the function of voltage-gated Ca2+ channels during prenatal development must be investigated in more detail, not least to understand devastative diseases like developmental epileptic encephalopathies (DEE).


Asunto(s)
Canales de Calcio Tipo R/fisiología , Canales de Calcio Tipo T/fisiología , Proteínas de Transporte de Catión/fisiología , Cromosomas/genética , Inestabilidad Genómica , Endogamia/métodos , Sitios de Carácter Cuantitativo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Yakugaku Zasshi ; 140(10): 1207-1212, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-32999199

RESUMEN

T-type calcium channels are low-threshold voltage-gated calcium channel and characterized by unique electrophysiological properties such as fast inactivation and slow deactivation kinetics. All subtypes of T-type calcium channel (Cav3.1, 3.2 and 3.3) are widely expressed in the central nerve system, and they have an important role in homeostasis of sleep, pain response, and development of epilepsy. Recently, several reports suggest that T-type calcium channels may mediate neuronal plasticity in the mouse brain. We succeeded to develop T-type calcium channel enhancer ethyl 8'-methyl-2',4-dioxo-2-(piperidin-1-yl)-2'H-spiro[cyclopentane-1,3'-imidazo[1,2-a]pyridine]-2-ene-3-carboxylate (SAK3) which enhances Cav3.1 and 3.3 currents in each-channel expressed neuro2A cells. SAK3 can promote acetylcholine (ACh) release in the mouse hippocampus via enhancing T-type calcium channel. In this review, we have introduced the role of T-type calcium channel, especially Cav3.1 channel in the mouse hippocampus based on our previous data using SAK3 and Cav3.1 knockout mice.


Asunto(s)
Canales de Calcio Tipo T/efectos de los fármacos , Canales de Calcio Tipo T/fisiología , Imidazoles/farmacología , Neuronas/fisiología , Compuestos de Espiro/farmacología , Acetilcolina/metabolismo , Animales , Encéfalo/fisiología , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Células Cultivadas , Sistema Nervioso Central/metabolismo , Fenómenos Electrofisiológicos , Epilepsia/etiología , Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Homeostasis , Ratones , Plasticidad Neuronal , Dolor/etiología , Ratas , Sueño/fisiología
12.
FEBS Open Bio ; 10(10): 2122-2136, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32865339

RESUMEN

Loss of T-type calcium channel (TCC) function has been reported to result in decreased cell viability and impaired muscle regeneration, but the underlying mechanisms remain largely unknown. We previously found that expression of TCC is reduced in aged pelvic floor muscle of multiple vaginal delivery mice, and this is related to endoplasmic reticulum stress (ERS) activation and autophagy flux blockade. In the present work, we further investigated the effects of TCC function loss on C2C12 myotubes and skeletal muscle, which is mediated by promotion of ERS and ultimately contributes to mitochondrial-related apoptotic cell death. We found that application of a TCC inhibitor induced mitochondria-related apoptosis in a dose-dependent manner and also reduced mitochondrial transmembrane potential (MMP), induced mito-ROS generation, and enhanced expression of mitochondrial apoptosis proteins. Functional inhibition of TCC induced ERS, resulting in disorder of Ca2+ homeostasis in endoplasmic reticulum, and ultimately leading to cell apoptosis in C2C12 myotubes. Tibialis anterior muscles of T-type α1H channel knockout mice displayed a smaller skeletal muscle fiber size and elevated ERS-mediated apoptosis signaling. Our data point to a novel mechanism whereby TCC blockade leads to ERS activation and terminal mitochondrial-related apoptotic events in C2C12 myotubes and skeletal muscles.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Canales de Calcio Tipo T/farmacología , Estrés del Retículo Endoplásmico/fisiología , Animales , Apoptosis/fisiología , Autofagia , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/fisiología , Línea Celular , China , Retículo Endoplásmico/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/fisiología , Proteínas Mitocondriales/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Especies Reactivas de Oxígeno , Transducción de Señal
13.
Bioelectrochemistry ; 136: 107618, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32795940

RESUMEN

A family of current-time curves of T-type Cav3.1 Ca2+ channels available in the literature is simulated by a kinetic model differing from that used for the interpretation of all salient features of Na+ and Shaker K+ channels by the insertion of a multiplying factor expressing the difference between the working potential ϕ and the reversal potential ϕr. This deterministic model is also used to simulate experimental curves taken from the literature for steady-state 'fast inactivation' and for a gradual passage from fast to 'slow inactivation'. A depolarizing pulse induces fast or slow inactivation depending on whether it lasts 100-500 ms or about 1 min, and is believed to cause a collapse of the central pore near the selectivity filter (SF). A number of features of fast and slow inactivation of Cav3.1 Ca2+ channels are qualitatively interpreted on the basis of a sequence of conformational states. Briefly, the conformation responsible for 'fast inactivation' is assumed to have the activation gate open and the inactivation gate (i.e., the SF) inactive. Immediately after a depolarizing pulse, this conformation is inactive and requires a sufficiently long rest time at a far negative holding potential to recover from inactivation. 'Slow inactivation' is ascribed to a different conformation with the activation gate closed and the SF inactive.


Asunto(s)
Canales de Calcio Tipo T/química , Modelos Moleculares , Canales de Calcio Tipo T/fisiología , Humanos , Activación del Canal Iónico , Conformación Proteica
14.
Life Sci ; 260: 118291, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-32810510

RESUMEN

AIMS: Despite the advanced cancer treatments, there is increased resistance to chemotherapy and subsequent mortality. In lack of reliable data in monolayer cultures and animal models, researchers are shifting to 3D cancer spheroids, which represents the in vivo robust tumour morphology. Calcium is essential in cell signalling and proliferation. It is found that T-type calcium channels (TTCCs) are overexpressed in various cancer cells, supporting their increased proliferation. Many of the TTCCs blockers available could target other channels besides TTCCs, which can cause adverse effects. Therefore, we hypothesise that TTA-A2, a highly selective blocker towards TTCCs, can inhibit the growth of cancer spheroids, and provide an anti-cancer and an adjuvant role in cancer therapy. METHODS: We studied TTA-A2 and paclitaxel (PTX-control drug) in lung adenocarcinoma cell line- A549, cancer cells and human embryonic kidney cell line- HEK 293, control cell, in their monolayer and spheroids forms for viability, proliferation, morphology change, migration, and invasion-after 48-96 h of treatment. KEY FINDINGS: Though the results varied between the monolayer and spheroids studies, we found both anti-cancer as well as adjuvant effect of TTA-A2 in both the studies. TTA-A2 was able to inhibit the growth, viability, and metastasis of the cancer cells and spheroids. Differences in the results of two modes might explain that why drugs tested successfully in monolayer culture fail in clinical trials. SIGNIFICANCE: This study establishes the role of TTA-A2, a potent TTCC blocker as an anti-cancer and adjuvant drug in reducing the viability and metastasis of the cancer cells.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Antineoplásicos , Bencenoacetamidas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/efectos de los fármacos , Neoplasias Pulmonares/patología , Piridinas/farmacología , Células A549 , Adenocarcinoma del Pulmón/tratamiento farmacológico , Bencenoacetamidas/uso terapéutico , Canales de Calcio Tipo T/fisiología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Invasividad Neoplásica/prevención & control , Piridinas/uso terapéutico
15.
J Neurosci ; 40(9): 1795-1809, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-31969470

RESUMEN

In mouse cerebellar Purkinje neurons (PNs), the climbing fiber (CF) input provides a signal to parallel fiber (PF) synapses, triggering PF synaptic plasticity. This signal is given by supralinear Ca2+ transients, associated with the CF synaptic potential and colocalized with the PF Ca2+ influx, occurring only when PF activity precedes the CF input. Here, we unravel the biophysical determinants of supralinear Ca2+ signals associated with paired PF-CF synaptic activity. We used membrane potential (Vm) and Ca2+ imaging to investigate the local CF-associated Ca2+ influx following a train of PF synaptic potentials in two cases: (1) when the dendritic Vm is hyperpolarized below the resting Vm, and (2) when the dendritic Vm is at rest. We found that supralinear Ca2+ signals are mediated by type-1 metabotropic glutamate receptors (mGluR1s) when the CF input is delayed by 100-150 ms from the first PF input in both cases. When the dendrite is hyperpolarized only, however, mGluR1s boost neighboring T-type channels, providing a mechanism for local coincident detection of PF-CF activity. The resulting Ca2+ elevation is locally amplified by saturation of endogenous Ca2+ buffers produced by the PF-associated Ca2+ influx via the mGluR1-mediated nonselective cation conductance. In contrast, when the dendritic Vm is at rest, mGluR1s increase dendritic excitability by inactivating A-type K+ channels, but this phenomenon is not restricted to the activated PF synapses. Thus, Vm is likely a crucial parameter in determining PF synaptic plasticity, and the occurrence of hyperpolarization episodes is expected to play an important role in motor learning.SIGNIFICANCE STATEMENT In Purkinje neurons, parallel fiber synaptic plasticity, determined by coincident activation of the climbing fiber input, underlies cerebellar learning. We unravel the biophysical mechanisms allowing the CF input to produce a local Ca2+ signal exclusively at the sites of activated parallel fibers. We show that when the membrane potential is hyperpolarized with respect to the resting membrane potential, type-1 metabotropic glutamate receptors locally enhance Ca2+ influx mediated by T-type Ca2+ channels, and that this signal is amplified by saturation of endogenous buffer also mediated by the same receptors. The combination of these two mechanisms is therefore capable of producing a Ca2+ signal at the activated parallel fiber sites, suggesting a role of Purkinje neuron membrane potential in cerebellar learning.


Asunto(s)
Señalización del Calcio/fisiología , Cerebelo/fisiología , Células de Purkinje/fisiología , Receptores AMPA/fisiología , Algoritmos , Animales , Canales de Calcio Tipo T/fisiología , Cerebelo/citología , Simulación por Computador , Dendritas/fisiología , Potenciales Postsinápticos Excitadores , Femenino , Masculino , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Plasticidad Neuronal/fisiología , Canales de Potasio/fisiología , Sinapsis/fisiología
16.
Neurosci Bull ; 36(5): 519-529, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31953800

RESUMEN

In the current study, we sought to investigate whether T-type Ca2+ channels (TCCs) in the brain are involved in generating post-anesthetic hyperexcitatory behaviors (PAHBs). We found that younger rat pups (postnatal days 9-11) had a higher incidence of PAHBs and higher PAHB scores than older pups (postnatal days 16-18) during emergence from sevoflurane anesthesia. The power spectrum of the theta oscillations (4 Hz-8 Hz) in the prefrontal cortex was significantly enhanced in younger pups when PAHBs occurred, while there were no significant changes in older pups. Both the power of theta oscillations and the level of PAHBs were significantly reduced by the administration of TCC inhibitors. Moreover, the sensitivity of TCCs in the medial dorsal thalamic nucleus to sevoflurane was found to increase with age by investigating the kinetic properties of TCCs in vitro. TCCs were activated by potentiated GABAergic depolarization with a sub-anesthetic dose of sevoflurane (1%). These data suggest that (1) TCCs in the brain contribute to the generation of PAHBs and the concomitant electroencephalographic changes; (2) the stronger inhibitory effect of sevoflurane contributes to the lack of PAHBs in older rats; and (3) the contribution of TCCs to PAHBs is not mediated by a direct effect of sevoflurane on TCCs.


Asunto(s)
Anestésicos por Inhalación/farmacología , Canales de Calcio Tipo T/fisiología , Locomoción/efectos de los fármacos , Sevoflurano/farmacología , Anestesia , Animales , Animales Recién Nacidos , Bloqueadores de los Canales de Calcio/farmacología , Electroencefalografía , Femenino , Masculino , Núcleo Talámico Mediodorsal/fisiología , Ratas , Ratas Sprague-Dawley , Ritmo Teta/fisiología
17.
Cereb Cortex ; 30(2): 488-504, 2020 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-31210267

RESUMEN

Neocortical GABAergic interneurons expressing vasoactive intestinal polypeptide (VIP) contribute to sensory processing, sensorimotor integration, and behavioral control. In contrast to other major subpopulations of GABAergic interneurons, VIP neurons show a remarkable diversity. Studying morphological and electrophysiological properties of VIP cells, we found a peculiar group of neurons in layer II/III of mouse primary somatosensory (barrel) cortex, which showed a highly dynamic burst firing behavior at resting membrane potential that switched to tonic mode at depolarized membrane potentials. Furthermore, we demonstrate that burst firing depends on T-type calcium channels. The burst-tonic switch could be induced by acetylcholine (ACh) and serotonin. ACh mediated a depolarization via nicotinic receptors whereas serotonin evoked a biphasic depolarization via ionotropic and metabotropic receptors in 48% of the population and a purely monophasic depolarization via metabotropic receptors in the remaining cells. These data disclose an electrophysiologically defined subpopulation of VIP neurons that via neuromodulator-induced changes in firing behavior is likely to regulate the state of cortical circuits in a profound manner.


Asunto(s)
Potenciales de Acción , Neuronas GABAérgicas/fisiología , Corteza Somatosensorial/fisiología , Péptido Intestinal Vasoactivo/análisis , Acetilcolina/administración & dosificación , Acetilcolina/fisiología , Animales , Canales de Calcio Tipo T/fisiología , Agonistas Colinérgicos/administración & dosificación , Neuronas GABAérgicas/efectos de los fármacos , Ratones Transgénicos , Serotonina/administración & dosificación , Serotonina/fisiología , Serotoninérgicos/administración & dosificación , Corteza Somatosensorial/diagnóstico por imagen
18.
Sci Signal ; 12(600)2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31551295

RESUMEN

Although brain-derived neurotrophic factor (BDNF) is implicated in the nociceptive signaling of peripheral sensory neurons, the underlying mechanisms remain largely unknown. Here, we elucidated the effects of BDNF on the neuronal excitability of trigeminal ganglion (TG) neurons and the pain sensitivity of rats mediated by T-type Ca2+ channels. BDNF reversibly and dose-dependently enhanced T-type channel currents through the activation of tropomyosin receptor kinase B (TrkB). Antagonism of phosphatidylinositol 3-kinase (PI3K) but not of its downstream target, the kinase AKT, abolished the BDNF-induced T-type channel response. BDNF application activated p38 mitogen-activated protein kinase (MAPK), and this effect was prevented by inhibition of PI3K but not of protein kinase A (PKA). Antagonism of either PI3K or p38 MAPK prevented the BDNF-induced stimulation of PKA activity, whereas PKA inhibition blocked the BDNF-mediated increase in T-type currents. BDNF increased the rate of action potential firing in TG neurons and enhanced the pain sensitivity of rats to mechanical stimuli. Moreover, inhibition of TrkB signaling abolished the increased mechanical sensitivity in a rat model of chronic inflammatory pain, and this effect was attenuated by either T-type channel blockade or knockdown of the channel Cav3.2. Together, our findings indicate that BDNF enhances T-type currents through the stimulation of TrkB coupled to PI3K-p38-PKA signaling, thereby inducing neuronal hyperexcitability of TG neurons and pain hypersensitivity in rats.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Canales de Calcio Tipo T/efectos de los fármacos , Dolor/fisiopatología , Células Receptoras Sensoriales/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Canales de Calcio Tipo T/metabolismo , Canales de Calcio Tipo T/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas Sprague-Dawley , Receptor trkB/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Transducción de Señal/efectos de los fármacos , Ganglio del Trigémino/citología , Ganglio del Trigémino/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Nihon Yakurigaku Zasshi ; 154(3): 97-102, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-31527367

RESUMEN

Among voltage-gated Ca2+ channels, T-type Ca2+ channels, which are activated by low voltages, regulate neuronal excitability, spontaneous neurotransmitter release, hormone secretion, etc. and also participate in proliferation of distinct cancer cells. Among three isoforms of T-type Ca2+ channels, Cav3.2 is detectable in 100% of biopsy samples from prostate cancer patients. In general, prostate cancer cells are highly sensitive to androgen deprivation therapy, but often acquire hormone-therapy resistance. The androgen deprivation may trigger neuroendocrine (NE)-like differentiation of some prostate cancer cells. We have analyzed the expression and function of Cav3.2 in human prostate cancer LNCaP cells during NE-like differentiation. NE-like LNCaP cells overexpress Cav3.2 through the CREB/Egr-1 pathway and also cystathionine-γ-lyase (CSE), which generates H2S that enhances the channel activity of Cav3.2. H2S generated by upregulated CSE appears to enhance the activity of upregulated Cav3.2 after the differentiation. The enhanced Cav3.2 activity in NE-like cells may contribute to increased secretion of mitogenic factors essential for androgen-independent proliferation of surrounding prostate cancer cells. It is known that increased extracellular glucose levels enhance Cav3.2 activity through asparagine (N)-linked glycosylation of Cav3.2, which might contribute to diabetic neuropathy. We then found that high glucose accelerates the enhanced channel function and overexpression of Cav3.2 in NE-like LNCaP cells, which might be associated with clinical evidence for diabetes-related poor prognosis of prostate cancer and development of hormone therapy resistance. Thus, Cav3.2 is considered to play a role in the pathophysiology of prostate cancer, and may serve as a therapeutic target.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Sistemas Neurosecretores/citología , Neoplasias de la Próstata/patología , Antagonistas de Andrógenos/farmacología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Cistationina gamma-Liasa/fisiología , Humanos , Sulfuro de Hidrógeno , Masculino
20.
J Therm Biol ; 84: 1-7, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31466741

RESUMEN

Heat can trigger testicular damage and impair fertility. Leydig cells produce testosterone in response to stimulation by luteinizing hormone (LH), which induces Ca2+ entry and K+ efflux through ion channels in their plasma membrane. Considering that mechanisms coordinating the Leydig cell responses to hyperthermic stress remain unclear; the present study analyzed the effects of heat stress (HS, 43°C, 15 min) and inhibition of Hsp90 on T-type calcium currents and voltage-dependent potassium currents (VKC) in mice Leydig cells. Results show that HS reduced the VKC steady state currents at +80 mV (45.3%) and maximum conductance (71.5%), as well as increased the activation time constant (31.7%) and the voltage for which half the channels are open (30%). Hsp90 inhibition did not change the VKC currents. T-type calcium currents were not affected by HS or Hsp90 inhibition. In conclusion, HS can slow the activation, reduce the currents and voltage dependence of the VKC, suggesting a possible role of these currents in the response to hyperthermic stress in Leydig cells.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Proteínas HSP90 de Choque Térmico/fisiología , Respuesta al Choque Térmico/fisiología , Células Intersticiales del Testículo/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Animales , Benzoquinonas/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Calor , Lactamas Macrocíclicas/farmacología , Células Intersticiales del Testículo/efectos de los fármacos , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...