Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Pflugers Arch ; 475(5): 595-606, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36964781

RESUMEN

The primary function of dystrophin is to form a link between the cytoskeleton and the extracellular matrix. In addition to this crucial structural function, dystrophin also plays an essential role in clustering and organizing several signaling proteins, including ion channels. Proteomic analysis of the whole rodent brain has stressed the role of some components of the dystrophin-associated glycoprotein complex (DGC) as potential interacting proteins of the voltage-gated Ca2+ channels of the CaV2 subfamily. The interaction of CaV2 with signaling and scaffolding proteins, such as the DGC components, may influence their function, stability, and location in neurons. This work aims to study the interaction between dystrophin and CaV2.1. Our immunoprecipitation data showed the presence of a complex formed by CaV2.1, CaVα2δ-1, CaVß4e, Dp140, and α1-syntrophin in the brain. Furthermore, proximity ligation assays (PLA) showed that CaV2.1 and CaVα2δ-1 interact with dystrophin in the hippocampus and cerebellum. Notably, Dp140 and α1-syntrophin increase CaV2.1 protein stability, half-life, permanence in the plasma membrane, and current density through recombinant CaV2.1 channels. Therefore, we have identified the Dp140 and α1-syntrophin as novel interaction partners of CaV2.1 channels in the mammalian brain. Consistent with previous findings, our work provides evidence of the role of DGC in anchoring and clustering CaV channels in a macromolecular complex.


Asunto(s)
Distrofina , Proteómica , Animales , Distrofina/genética , Distrofina/metabolismo , Mamíferos/metabolismo , Neuronas/metabolismo
2.
Pflugers Arch ; 474(4): 457-468, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35235008

RESUMEN

Neuropathic pain is one of the primary forms of chronic pain and is the consequence of the somatosensory system's direct injury or disease. It is a relevant public health problem that affects about 10% of the world's general population. In neuropathic pain, alteration in neurotransmission occurs at various levels, including the dorsal root ganglia, the spinal cord, and the brain, resulting from the malfunction of diverse molecules such as receptors, ion channels, and elements of specific intracellular signaling pathways. In this context, there have been exciting advances in elucidating neuropathic pain's cellular and molecular mechanisms in the last decade, including the possible role that long non-coding RNAs (lncRNAs) may play, which open up new alternatives for the development of diagnostic and therapeutic strategies for this condition. This review focuses on recent studies associated with the possible relevance of lncRNAs in the development and maintenance of neuropathic pain through their actions on the functional expression of ion channels. Recognizing the changes in the function and spatio-temporal patterns of expression of these membrane proteins is crucial to understanding the control of neuronal excitability in chronic pain syndromes.


Asunto(s)
Dolor Crónico , Neuralgia , ARN Largo no Codificante , Animales , Dolor Crónico/genética , Modelos Animales de Enfermedad , Ganglios Espinales/metabolismo , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Neuralgia/genética , Neuralgia/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
3.
J Neurophysiol ; 128(6): 1555-1564, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36350063

RESUMEN

Neuronal L-type Ca2+ channels of the CaV1.3 subclass are transmembrane protein complexes that contribute to the pacemaker activity in the adult substantia nigra dopaminergic neurons. The altered function of these channels may play a role in the development and progress of neurodegenerative mechanisms implicated in Parkinson's disease (PD). Although L-type channel expression is precisely regulated, an increased functional expression has been observed in PD. Previously, we showed that Parkin, an E3 enzyme of the ubiquitin-proteasome system (UPS) interacts with neuronal CaV2.2 channels promoting their ubiquitin-mediated degradation. In addition, previous studies show an increase in CaV1.3 channel activity in dopaminergic neurons of the SNc and that Parkin expression is reduced in PD. These findings suggest that the decrease in Parkin may affect the proteasomal degradation of CaV1.3, which helps explain the increase in channel activity. Therefore, the present report aims to gain insight into the degradation mechanisms of the neuronal CaV1.3 channel by the UPS. Immunoprecipitation assays showed the interaction between Parkin and the CaV1.3 channels expressed in HEK-293 cells and neural tissues. Likewise, Parkin overexpression reduced the total and membrane channel levels and decreased the current density. Consistent with this, patch-clamp recordings in the presence of an inhibitor of the UPS, MG132, prevented the effects of Parkin, suggesting enhanced channel proteasomal degradation. In addition, the half-life of the pore-forming CaV1.3α1 protein was significantly reduced by Parkin overexpression. Finally, electrophysiological recordings using a PRKN knockout HEK-293 cell line generated by CRISPR/Cas9 showed increased current density. These results suggest that Parkin promotes the proteasomal degradation of CaV1.3, which may be a relevant aspect for the pathophysiology of PD.NEW & NOTEWORTHY The increased expression of CaV1.3 calcium channels is a crucial feature of Parkinson's disease (PD) pathophysiology. However, the mechanisms that determine this increase are not yet defined. Parkin, an enzyme of the ubiquitin-proteasome system, is known to interact with neuronal channels promoting their ubiquitin-mediated degradation. Interestingly, Parkin mutations also play a role in PD. Here, the degradation mechanisms of CaV1.3 channels and their relationship with the pathophysiology of PD are studied in detail.


Asunto(s)
Canales de Calcio Tipo L , Enfermedad de Parkinson , Ubiquitina-Proteína Ligasas , Humanos , Neuronas Dopaminérgicas/metabolismo , Células HEK293 , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo
4.
Int J Neurosci ; : 1-10, 2022 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-35993158

RESUMEN

Aim: Voltage-gated calcium (CaV) channels play an essential role in maintaining calcium homeostasis and regulating numerous physiological processes in neurons. Therefore, dysregulation of calcium signaling is relevant in many neurological disorders, including Parkinson's disease (PD). This review aims to introduce the role of CaV channels in PD and discuss some novel aspects of channel regulation and its impact on the molecular pathophysiology of the disease.Methods: an exhaustive search of the literature in the field was carried out using the PubMed database of The National Center for Biotechnology Information. Systematic searches were performed from the initial date of publication to May 2022.Results: Although α-synuclein aggregates are the main feature of PD, L-type calcium (CaV1) channels seem to play an essential role in the pathogenesis of PD. Changes in the functional expression of CaV1.3 channels alter Calcium homeostasis and contribute to the degeneration of dopaminergic neurons. Furthermore, recent studies suggest that CaV channel trafficking towards the cell membrane depends on the activity of the ubiquitin-proteasome system (UPS). In PD, there is an increase in the expression of L-type channels associated with a decrease in the expression of Parkin, an E3 enzyme of the UPS. Therefore, a link between Parkin and CaV channels could play a fundamental role in the pathogenesis of PD and, as such, could be a potentially attractive target for therapeutic intervention.Conclusion: The study of alterations in the functional expression of CaV channels will provide a framework to understand better the neurodegenerative processes that occur in PD and a possible path toward identifying new therapeutic targets to treat this condition.

5.
Int J Mol Sci ; 23(15)2022 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-35897646

RESUMEN

The CatSper channel localizes exclusively in the flagella of sperm cells. The Catsper1 protein, together with three pore units, is essential for the CatSper Channel formation, which produces flagellum hyperactivation and confers sperm fertility. Catsper1 expression is dependent on Sox transcription factors, which can recognize in vitro at least three Sox binding sites on the promoter. Sox transcription factors have calmodulin-binding domains for nuclear importation. Calmodulin (CaM) is affected by the specific inhibitor calmidazolium (CMZ), which prevents the nuclear transport of Sox factors. In this work, we assess the regulation of the Catsper1 promoter in vivo by Sox factors in the murine testis and evaluate the effects of the inhibitor calmidazolium on the expression of the Casper genes, and the motility and fertility of the sperm. Catsper1 promoter has significant transcriptional activity in vivo; on the contrary, three Sox site mutants in the Catsper1 promoter reduced transcriptional activity in the testis. CaM inhibition affects Sox factor nuclear transport and has notable implications in the expression and production of Catsper1, as well as in the motility and fertility capability of sperm. The molecular mechanism described here might conform to the basis of a male contraceptive strategy acting at the transcriptional level by affecting the production of the CatSper channel, a fundamental piece of male fertility.


Asunto(s)
Canales de Calcio , Calmodulina , Animales , Canales de Calcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Regulación hacia Abajo , Fertilidad , Imidazoles , Masculino , Ratones , Factores de Transcripción SOX/genética , Semen/metabolismo , Motilidad Espermática/fisiología , Espermatozoides/metabolismo
6.
J Neurosci ; 40(2): 283-296, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31744861

RESUMEN

Voltage-gated T-type Ca2+ (CaV3) channels regulate diverse physiological events, including neuronal excitability, and have been linked to several pathological conditions such as absence epilepsy, cardiovascular diseases, and neuropathic pain. It is also acknowledged that calcium/calmodulin-dependent protein kinase II and protein kinases A and C regulate the activity of T-type channels. Interestingly, peripheral nerve injury induces tactile allodynia and upregulates CaV3.2 channels and cyclin-dependent kinase 5 (Cdk5) in dorsal root ganglia (DRG) and spinal dorsal horn. Here, we report that recombinant CaV3.2 channels expressed in HEK293 cells are regulatory targets of Cdk5. Site-directed mutagenesis showed that the relevant sites for this regulation are residues S561 and S1987. We also found that Cdk5 may regulate CaV3.2 channel functional expression in rats with mechanical allodynia induced by spinal nerve ligation (SNL). Consequently, the Cdk5 inhibitor olomoucine affected the compound action potential recorded in the spinal nerves, as well as the paw withdrawal threshold. Likewise, Cdk5 expression was upregulated after SNL in the DRG. These findings unveil a novel mechanism for how phosphorylation may regulate CaV3.2 channels and suggest that increased channel activity by Cdk5-mediated phosphorylation after SNL contributes nerve injury-induced tactile allodynia.SIGNIFICANCE STATEMENT Neuropathic pain is a current public health challenge. It can develop as a result of injury or nerve illness. It is acknowledged that the expression of various ion channels can be altered in neuropathic pain, including T-type Ca2+ channels that are expressed in sensory neurons, where they play a role in the regulation of cellular excitability. The present work shows that the exacerbated expression of Cdk5 in a preclinical model of neuropathic pain increases the functional expression of CaV3.2 channels. This finding is relevant for the understanding of the molecular pathophysiology of the disease. Additionally, this work may have a substantial translational impact, since it describes a novel molecular pathway that could represent an interesting therapeutic alternative for neuropathic pain.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Potenciales de Acción/fisiología , Animales , Células HEK293 , Humanos , Ligadura , Masculino , Traumatismos de los Nervios Periféricos/metabolismo , Fosforilación , Ratas , Ratas Wistar , Nervios Espinales/lesiones , Nervios Espinales/cirugía
7.
Am J Physiol Endocrinol Metab ; 319(1): E232-E244, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32369417

RESUMEN

Voltage-gated Ca2+ (CaV) channels are expressed in endocrine cells where they contribute to hormone secretion. Diverse chemical messengers, including epidermal growth factor (EGF), are known to affect the expression of CaV channels. Previous studies have shown that EGF increases Ca2+ currents in GH3 pituitary cells by increasing the number of high voltage-activated (HVA) CaV channels at the cell membrane, which results in enhanced prolactin (PRL) secretion. However, little is known regarding the mechanisms underlying this regulation. Here, we show that EGF actually increases the expression of the CaVα2δ-1 subunit, a key molecular component of HVA channels. The analysis of the gene promoter encoding CaVα2δ-1 (CACNA2D1) revealed binding sites for transcription factors activated by the Ras/Raf/MEK/ERK signaling cascade. Chromatin immunoprecipitation and site-directed mutagenesis showed that ELK-1 is crucial for the transcriptional regulation of CACNA2D1 in response to EGF. Furthermore, we found that EGF increases the membrane expression of CaVα2δ-1 and that ELK-1 overexpression increases HVA current density, whereas ELK-1 knockdown decreases the functional expression of the channels. Hormone release assays revealed that CaVα2δ-1 overexpression increases PRL secretion. These results suggest a mechanism for how EGF, by activating the Ras/Raf/MEK/ERK/ELK-1 pathway, may influence the expression of HVA channels and the secretory behavior of pituitary cells.


Asunto(s)
Canales de Calcio Tipo L/genética , Factor de Crecimiento Epidérmico/metabolismo , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas/genética , Proteína Elk-1 con Dominio ets/genética , Quinasas raf/genética , Proteínas ras/genética , Animales , Canales de Calcio Tipo L/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Técnicas de Silenciamiento del Gen , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Proteína Elk-1 con Dominio ets/metabolismo , Quinasas raf/metabolismo , Proteínas ras/metabolismo
8.
Biochem Biophys Res Commun ; 524(1): 255-261, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-31983427

RESUMEN

Neurotransmission is one of the most important processes in neuronal communication and depends largely on Ca2+ entering synaptic terminals through voltage-gated Ca2+ (CaV) channels. Although the contribution of L-type CaV channels in neurotransmission has not been unambiguously established, increasing evidence suggests a role for these proteins in noradrenaline, dopamine, and GABA release. Here we report the regulation of L-type channels by Cdk5, and its possible effect on GABA release in the substantia nigra pars reticulata (SNpr). Using patch-clamp electrophysiology, we show that Cdk5 inhibition by Olomoucine significantly increases current density through CaV1.3 (L-type) channels heterologously expressed in HEK293 cells. Likewise, in vitro phosphorylation showed that Cdk5 phosphorylates residue S1947 in the C-terminal region of the pore-forming subunit of CaV1.3 channels. Consistent with this, the mutation of serine into alanine (S1947A) prevented the regulation of Cdk5 on CaV1.3 channel activity. Our data also revealed that the inhibition of Cdk5 increased the frequency of high K+-evoked miniature inhibitory postsynaptic currents in rat SNpr neurons, acting on L-type channels. These results unveil a novel regulatory mechanism of GABA release in the SNpr that involves a direct action of Cdk5 on L-type channels.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Potenciales Postsinápticos Inhibidores , Neostriado/metabolismo , Receptores de GABA-A/metabolismo , Sustancia Negra/metabolismo , Animales , Animales Recién Nacidos , Canales de Calcio Tipo L/química , Células HEK293 , Humanos , Masculino , Fosforilación , Ratas Wistar , Ácido gamma-Aminobutírico/metabolismo
9.
Oncology ; 97(6): 373-382, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31430760

RESUMEN

INTRODUCTION: Breast cancer is one of the leading causes of death worldwide and is the result of dysregulation of various signaling pathways in mammary epithelial cells. The mortality rate in patients suffering from breast cancer is high because the tumor cells have a prominent invasive capacity towards the surrounding tissues. Previous studies carried out in tumor cell models show that voltage-gated ion channels may be important molecular actors that contribute to the migratory and invasive capacity of the tumor cells. METHODS: In this study, by using an experimental strategy that combines cell and molecular biology assays with electrophysiological recording, we sought to determine whether the voltage-dependent sodium channel NaV1.5 regulates the migratory capacity of the human breast cancer cell line MDA-MB 231, when cells are maintained in the presence of epidermal growth factor (EGF), as an inductor of the epithelial-mesenchymal transition. RESULTS: Our data show that EGF stimulates the migratory capacity of MDA-MB 231 cells, by regulating the functional expression of NaV1.5 channels. Consistent with this, the stimulatory actions of the growth factor were prevented by the use of tetrodotoxin, an Na+ channel selective blocker, as well as by resveratrol, an antioxidant that can also affect Na+ channel activity. DISCUSSION: The understanding of molecular mechanisms, such as the EGF pathway in the progression of breast cancer is fundamental for the design of more effective therapeutic strategies for the disease.


Asunto(s)
Neoplasias de la Mama/patología , Factor de Crecimiento Epidérmico/farmacología , Canal de Sodio Activado por Voltaje NAV1.5/fisiología , Calcio/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal , Femenino , Humanos , Canal de Sodio Activado por Voltaje NAV1.5/análisis , Resveratrol/farmacología
10.
J Membr Biol ; 251(4): 535-550, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29779049

RESUMEN

Dystrophin is a cytoskeleton-linked membrane protein that binds to a larger multiprotein assembly called the dystrophin-associated glycoprotein complex (DGC). The deficiency of dystrophin or the components of the DGC results in the loss of connection between the cytoskeleton and the extracellular matrix with significant pathophysiological implications in skeletal and cardiac muscle as well as in the nervous system. Although the DGC plays an important role in maintaining membrane stability, it can also be considered as a versatile and flexible molecular complex that contribute to the cellular organization and dynamics of a variety of proteins at specific locations in the plasma membrane. This review deals with the role of the DGC in transmembrane signaling by forming supramolecular assemblies for regulating ion channel localization and activity. These interactions are relevant for cell homeostasis, and its alterations may play a significant role in the etiology and pathogenesis of various disorders affecting muscle and nerve function.


Asunto(s)
Distrofina/metabolismo , Glicoproteínas/metabolismo , Canales Iónicos/metabolismo , Animales , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Transducción de Señal
11.
Biochem Biophys Res Commun ; 491(1): 53-58, 2017 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-28705737

RESUMEN

Leptin, a peptide hormone produced by adipocytes, is recognized as one of the signals involved in the onset of reproductive activity. The leptin receptor has been found in hypothalamic neurons and pituitary gonadotropes, suggesting that the hormone may act at both sites to stimulate the secretion of GnRH and consequently, FSH and LH. In response to a stimulus such as a hypothalamic secretagogue, gonadotropes respond with changes in electrical activity, intracellular Ca2+ and hormone release. The main aim of this report was to investigate whether leptin promotes a change in the electrical and secretory activities of bovine gonadotropes. After 48 h of treatment with leptin (10 nM) significant changes in the action potential properties were observed in gonadotropes, which included an increase in amplitude, time-to-pike and post-hyperpolarization, as well as a decrease in firing threshold. Likewise, leptin induced a significant (∼1.3-fold) up-regulation of voltage-gated Na+ channel current density, and a selective increase (∼2.1-fold) in Ca2+ current density through high voltage-activated channels. Consistent with this, leptin enhanced GnRH-induced secretion of LH measured by ELISA. We suggest that leptin enhances membrane expression of voltage-gated Na+ and Ca2+ channels, which results in a modulation of the action potential properties and an increase in hormone release from gonadotropes.


Asunto(s)
Potenciales de Acción/fisiología , Células Endocrinas/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Leptina/metabolismo , Hormona Luteinizante/metabolismo , Potenciales de la Membrana/fisiología , Animales , Bovinos , Células Cultivadas , Masculino
12.
Oncology ; 93(1): 1-10, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28355609

RESUMEN

Extensive research is currently underway, seeking better diagnostic methods and treatments and a better understanding of the molecular mechanisms involved in cancer, from the role of specific genetic mutations to the intricate biochemical and molecular pathways involved. Because of their role in regulating relevant physiological events such as cell proliferation, migration, and invasion, ion channels have recently been recognized as important elements in cancer initiation and progression. Moreover, it has been reported that pharmacological intervention in ion channel activity might provide protection against diverse types of cancer, and that ion channels could be used as targets to counteract tumor growth, prevent metastasis, and overcome the therapy resistance of tumor cells. In this context, Ca2+ channels have been found to play a role in tumorigenesis and tumor progression. Specifically, L-type Ca2+ channel inhibition may affect cell proliferation, differentiation, and apoptosis. This review aims to provide insights into the potential role of these channels in cancer cell lines, emphasizing their participation in cell proliferation, migration, and autophagy induction, as well as their potential as rational targets for new cancer therapeutics.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Movimiento Celular , Proliferación Celular , Neoplasias/genética , Neoplasias/patología , Autofagia , Canales de Calcio Tipo L/genética , Señalización del Calcio/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico
13.
Gen Physiol Biophys ; 36(1): 1-5, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27787228

RESUMEN

Recent findings have revealed a fundamental role of the ubiquitin-proteasome system (UPS) in the regulation of voltage-gated Ca2+ channels (VGCCs). It has been proposed that the ubiquitination-deubiquitination balance dictates the number of channels expressed at the plasma membrane, which in turn influences a number of physiological and pathophysiological processes. This minireview surveys recent studies showing that VGCCs may be ubiquitinated in an unexpectedly complex manner, and highlights the role of the UPS in the regulation of the channels, focusing on the mechanisms that control their cell surface expression. The exciting new findings in this emerging field suggest that the turnover of VGCCs may be determined to a large degree by the activity of the UPS, and that alteration of the UPS molecular machinery may be one of the underlying mechanisms occurring in a number of channelopathies.


Asunto(s)
Calcio/metabolismo , Canalopatías/metabolismo , Activación del Canal Iónico , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación , Ubiquitinas/metabolismo , Animales , Señalización del Calcio , Humanos , Modelos Biológicos , Proteínas Ubiquitinadas/metabolismo
14.
J Cell Physiol ; 229(5): 551-60, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23868804

RESUMEN

Voltage-gated T-type Ca(2+) (CaV 3) channels mediate a number of physiological events in developing and mature cells, and are implicated in neurological and cardiovascular diseases. In mammals, there are three distinct T-channel genes (CACNA1G, CACNA1H, and CACNA1I) encoding proteins (CaV 3.1-CaV 3.3) that differ in their localization as well as in molecular, biophysical, and pharmacological properties. The CACNA1G is a large gene that contains 38 exons and is localized in chromosome 17q22. Only basic characteristics of the CACNA1G gene promoter region have been investigated classifying it as a TATA-less sequence containing several potential transcription factor-binding motifs. Here, we cloned and characterized a proximal promoter region and initiated the analysis of transcription factors that control CaV 3.1 channel expression using the murine Cacna1g gene as a model. We isolated a ∼1.5 kb 5'-upstream region of Cacna1g and verified its transcriptional activity in the mouse neuroblastoma N1E-115 cell line. In silico analysis revealed that this region possesses a TATA-less minimal promoter that includes two potential transcription start sites and four binding sites for the transcription factor Sp1. The ability of one of these sites to interact with the transcription factor was confirmed by electrophoretic mobility shift assays. Consistent with this, Sp1 over-expression enhanced promoter activity while siRNA-mediated Sp1 silencing significantly decreased the level of CaV 3.1 protein and reduced the amplitude of whole-cell T-type Ca(2+) currents expressed in the N1E-115 cells. These results provide new insights into the molecular mechanisms that control CaV 3.1 channel expression.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Factor de Transcripción Sp1/metabolismo , Animales , Secuencia de Bases , Canales de Calcio Tipo T/genética , Línea Celular , Clonación Molecular , Regulación de la Expresión Génica , Silenciador del Gen , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/genética , Canales Aniónicos Dependientes del Voltaje
15.
Pflugers Arch ; 466(11): 2113-26, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24566975

RESUMEN

Microtubule-associated protein B is a cytoskeleton protein consisting of heavy and light (LC) chains that play important roles in the regulation of neuronal morphogenesis and function. LC1 is also well known to interact with diverse ionotropic receptors at postsynapse. Much less is known, however, regarding the role of LC1 at presynaptic level where voltage-gated N-type Ca(2+) channels couple membrane depolarization to neurotransmitter release. Here, we investigated whether LC1 interacts with the N-type channels. Co-localization analysis revealed spatial proximity of the two proteins in hippocampal neurons. The interaction between LC1 and the N-type channel was demonstrated using co-immunoprecipitation experiments and in vitro pull-down assays. Detailed biochemical analysis suggested that the interaction occurs through the N-terminal of LC1 and the C-terminal of the pore-forming CaVα1 subunit of the channels. Patch-clamp studies in HEK-293 cells revealed a significant decrease in N-type currents upon LC1 expression, without apparent changes in kinetics. Recordings performed in the presence of MG132 prevented the actions of LC1 suggesting enhanced channel proteasomal degradation. Interestingly, using the yeast two-hybrid system and immunoprecipitation assays in HEK-293 cells, we revealed an interaction between LC1 and the ubiquitin-conjugating enzyme UBE2L3. Furthermore, we found that the LC1/UBE2L3 complex could interact with the N-type channels, suggesting that LC1 may act as a scaffold protein to increase UBE2L3-mediated channel ubiquitination. Together these results revealed a novel functional coupling between LC1 and the N-type channels.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Membrana Celular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación/fisiología , Células Cultivadas , Células HEK293 , Hipocampo/metabolismo , Humanos , Inmunoprecipitación/métodos , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Técnicas del Sistema de Dos Híbridos , Ubiquitina/metabolismo
16.
Bioorg Med Chem ; 22(6): 1797-803, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24582401

RESUMEN

Neuropathic pain is a serious physical disabling condition resulting from lesion or dysfunction of the peripheral sensory nervous system. Despite the fact that the mechanisms underlying neuropathic pain are poorly understood, the involvement of voltage-gated calcium (Ca(V)) channels in its pathophysiology has justified the use of drugs that bind the Ca(V) channel α2δ auxiliary subunit, such as gabapentin (GBP), to attain analgesic and anti-allodynic effects in models involving neuronal sensitization and nerve injury. GBP binding to α2δ inhibits nerve injury-induced trafficking of the α1 pore forming subunits of Ca(V) channels, particularly of the N-type, from the cytoplasm to the plasma membrane of pre-synaptic terminals in dorsal root ganglion neurons and dorsal horn spinal neurons. In the search for alternative forms of treatment, in this study we describe the synthesis and pharmacological profile of a GABA derivative, 2-aminoadamantane-1-carboxylic acid (GZ4), which displays a close structure-activity relationship with GBP. Behavioral assessment using von Frey filament stimuli showed that GZ4 treatment reverted mechanical allodynia/hyperalgesia in an animal model of spinal nerve ligation-induced neuropathic pain. In addition, using the patch clamp technique we show that GZ4 treatment significantly decreased whole-cell currents through N-type Ca(V) channels heterologously expressed in HEK-293 cells. Interestingly, the behavioral and electrophysiological time course of GZ4 actions reflects that its mechanism of action is similar but not identical to that of GBP. While GBP actions require at least 24 h and imply uptake of the drug, which suggests that the drug acts mainly intracellularly affecting channels trafficking to the plasma membrane, the faster time course (1-3 h) of GZ4 effects suggests also a direct inhibition of Ca(2+) currents acting on cell surface channels.


Asunto(s)
Adamantano/análogos & derivados , Analgésicos/farmacología , Canales de Calcio Tipo N/metabolismo , Neuralgia/tratamiento farmacológico , Adamantano/síntesis química , Adamantano/química , Adamantano/farmacología , Analgésicos/síntesis química , Analgésicos/química , Analgésicos/uso terapéutico , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
17.
Biomedicines ; 12(1)2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38255311

RESUMEN

The D1R and D3R receptors functionally and synergistically interact in striatonigral neurons. Dopaminergic denervation turns this interaction antagonistic, which is correlated with a decrement in D3nf isoform and an increment in D3R membranal expression. The mechanisms of such changes in D3R are attributed to the dysregulation of the expression of their isoforms. The cause and mechanism of this phenomenon remain unknown. Dopaminergic denervation produces a decrement in D1R and PKA activity; we propose that the lack of phosphorylation of PTB (regulator of alternative splicing) by PKA produces the dysregulation of D3R splicing and changes D3R functionality. By using in silico analysis, we found that D3R mRNA has motifs for PTB binding and, by RIP, co-precipitates with PTB. Moreover, D1R activation via PKA promotes PTB phosphorylation. Acute and 5-day D1R blockade decreases the expression of D3nf mRNA. The 5-day treatment reduces D3R, D3nf, and PTB protein in the cytoplasm and increases D3R in the membrane and PTB in the nucleus. Finally, the blockade of D1R mimics the effect of dopaminergic denervation in D1R and D3R signaling. Thus, our data indicate that through PKA→PTB, D1R modulates D3R splicing, expression, and signaling, which are altered during D1R blockade or the lack of stimulation in dopaminergic denervation.

18.
Biochim Biophys Acta ; 1818(3): 551-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22198390

RESUMEN

Stimulus-secretion coupling is a complex set of intracellular reactions initiated by an external stimulus that result in the release of hormones and neurotransmitters. Under physiological conditions this signaling process takes a few milliseconds, and to minimize delays cells have developed a formidable integrated network, in which the relevant molecules are tightly packed on the nanometer scale. Active zones, the sites of release, are composed of several different proteins including voltage-gated Ca(2+) (Ca(V)) channels. It is well acknowledged that hormone and neurotransmitter release is initiated by the activation of these channels located close to docked vesicles, though the mechanisms that enrich channels at release sites are largely unknown. Interestingly, Rab3 binding proteins (RIMs), a diverse multidomain family of proteins that operate as effectors of the small G protein Rab3 involved in secretory vesicle trafficking, have recently identified as binding partners of Ca(V) channels, placing both proteins in the center of an interaction network in the molecular anatomy of the active zones that influence different aspects of secretion. Here, we review recent evidences providing support for the notion that RIMs directly bind to the pore-forming and auxiliary ß subunits of Ca(V) channels and with RIM-binding protein, another interactor of the channels. Through these interactions, RIMs regulate the biophysical properties of the channels and their anchoring relative to active zones, significantly influencing hormone and neurotransmitter release.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Neurotransmisores/metabolismo , Vesículas Secretoras/metabolismo , Proteínas de Unión al GTP rab3/metabolismo , Animales , Transporte Biológico Activo/fisiología , Humanos
19.
Biochim Biophys Acta ; 1822(8): 1238-46, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22549042

RESUMEN

Familial hemiplegic migraine type 1 (FHM-1) is a monogenic form of migraine with aura that is characterized by recurrent attacks of a typical migraine headache with transient hemiparesis during the aura phase. In a subset of patients, additional symptoms such as epilepsy and cerebellar ataxia are part of the clinical phenotype. FHM-1 is caused by missense mutations in the CACNA1A gene that encodes the pore-forming subunit of Ca(V)2.1 voltage-gated Ca(2+) channels. Although the functional effects of an increasing number of FHM-1 mutations have been characterized, knowledge on the influence of most of these mutations on G protein regulation of channel function is lacking. Here, we explored the effects of G protein-dependent modulation on mutations W1684R and V1696I which cause FHM-1 with and without cerebellar ataxia, respectively. Both mutations were introduced into the human Ca(V)2.1α(1) subunit and their functional consequences investigated after heterologous expression in human embryonic kidney 293 (HEK-293) cells using patch-clamp recordings. When co-expressed along with the human µ-opioid receptor, application of the agonist [d-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) inhibited currents through both wild-type (WT) and mutant Ca(V)2.1 channels, which is consistent with the known modulation of these channels by G protein-coupled receptors. Prepulse facilitation, which is a way to characterize the relief of direct voltage-dependent G protein regulation, was reduced by both FHM-1 mutations. Moreover, the kinetic analysis of the onset and decay of facilitation showed that the W1684R and V1696I mutations affect the apparent dissociation and reassociation rates of the Gßγ dimer from the channel complex, suggesting that the G protein-Ca(2+) channel affinity may be altered by the mutations. These biophysical studies may shed new light on the pathophysiology underlying FHM-1.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/metabolismo , Proteínas de Unión al GTP/metabolismo , Trastornos Migrañosos/genética , Trastornos Migrañosos/metabolismo , Animales , Canales de Calcio Tipo N/genética , Línea Celular , Proteínas de Unión al GTP/genética , Estudio de Asociación del Genoma Completo , Genotipo , Células HEK293 , Humanos , Activación del Canal Iónico , Ratones , Mutación , Ratas , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Transfección
20.
Pflugers Arch ; 465(6): 819-28, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23242029

RESUMEN

The α2δ proteins are auxiliary subunits of high-voltage-activated Ca(2+) channels associated with alterations of surface expression, kinetics, and voltage-dependent properties of the channel complex. Four mammalian genes and several splice α2δ subunit variants have been cloned and described, though very little information concerning the transcriptional mechanisms that regulate their expression is available. Here, we report the identification and characterization of the human α2δ-1 subunit gene promoter and its regulation by specific transcription factor 1 (Sp1). Transient transfection of human neuroblastoma SH-SY5Y cells with a promoter/luciferase reporter construct revealed a ~1.5 kb 5´-UTR fragment of the CACNA2D1 gene that produced high levels of luciferase activity. Deletional analysis of this sequence showed that the minimal promoter was located within a 413-bp region (nt -326 to +98) with respect to the transcription start site. In this region, no canonical TATA box was present, but a high GC content and five potential Sp1 binding sites were found. The ability of two of these sites to interact with the transcription factor was confirmed by electrophoretic mobility shift assays. Likewise, Sp1 overexpression enhanced promoter activity while siRNA-mediated Sp1 silencing significantly decreased the level of α2δ protein expressed in the SH-SY5Y cells, as well as reduced the amplitude of whole-cell patch clamp Ca(2+) currents in dorsal root ganglion neurons. This study thus represents the first identification of the transcriptional control region in the gene encoding the Ca(2+) channel α2δ-1 auxiliary subunit.


Asunto(s)
Regiones no Traducidas 5' , Canales de Calcio/genética , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/metabolismo , Potenciales de Acción , Animales , Composición de Base , Secuencia de Bases , Sitios de Unión , Calcio/metabolismo , Canales de Calcio/química , Canales de Calcio/metabolismo , Línea Celular Tumoral , Células Cultivadas , Ganglios Espinales/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neuronas/metabolismo , Neuronas/fisiología , Análisis de Secuencia de ADN , Factor de Transcripción Sp1/genética , TATA Box , Sitio de Iniciación de la Transcripción , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA