Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Biol Chem ; 297(4): 101126, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34461097

RESUMEN

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


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Calmodulina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Canales Catiónicos TRPC/metabolismo , Vacuolas/metabolismo , Calcio/química , Calmodulina/antagonistas & inhibidores , Calmodulina/química , Calmodulina/genética , Células HEK293 , Humanos , Dominios Proteicos , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Sesterterpenos/farmacología , Canales Catiónicos TRPC/química , Canales Catiónicos TRPC/genética , Vacuolas/química , Vacuolas/genética
2.
Int J Mol Sci ; 22(23)2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34884497

RESUMEN

Recently, we reported a case of an infant with neonatal severe under-mineralizing skeletal dysplasia caused by mutations within both alleles of the TRPV6 gene. One mutation results in an in frame stop codon (R510stop) that leads to a truncated, nonfunctional TRPV6 channel, and the second in a point mutation (G660R) that, surprisingly, does not affect the Ca2+ permeability of TRPV6. We mimicked the subunit composition of the unaffected heterozygous parent and child by coexpressing the TRPV6 G660R and R510stop mutants and combinations with wild type TRPV6. We show that both the G660R and R510stop mutant subunits are expressed and result in decreased calcium uptake, which is the result of the reduced abundancy of functional TRPV6 channels within the plasma membrane. We compared the proteomic profiles of a healthy placenta with that of the diseased infant and detected, exclusively in the latter two proteases, HTRA1 and cathepsin G. Our results implicate that the combination of the two mutant TRPV6 subunits, which are expressed in the placenta of the diseased child, is responsible for the decreased calcium uptake, which could explain the skeletal dysplasia. In addition, placental calcium deficiency also appears to be associated with an increase in the expression of proteases.


Asunto(s)
Canales de Calcio/genética , Catepsina G/metabolismo , Serina Peptidasa A1 que Requiere Temperaturas Altas/metabolismo , Mutación , Osteocondrodisplasias/patología , Placenta/patología , Proteoma/metabolismo , Canales Catiónicos TRPV/genética , Secuencia de Aminoácidos , Animales , Canales de Calcio/metabolismo , Canales de Calcio/fisiología , Estudios de Casos y Controles , Catepsina G/genética , Femenino , Regulación Enzimológica de la Expresión Génica , Serina Peptidasa A1 que Requiere Temperaturas Altas/genética , Humanos , Lactante , Ratones Noqueados , Osteocondrodisplasias/etiología , Osteocondrodisplasias/metabolismo , Placenta/metabolismo , Embarazo , Proteoma/análisis , Canales Catiónicos TRPV/metabolismo , Canales Catiónicos TRPV/fisiología
3.
Int J Mol Sci ; 21(24)2020 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-33352987

RESUMEN

Calcium-selective transient receptor potential Vanilloid 6 (TRPV6) channels are expressed in fetal labyrinth trophoblasts as part of the feto-maternal barrier, necessary for sufficient calcium supply, embryo growth, and bone development during pregnancy. Recently, we have shown a less- compact labyrinth morphology of Trpv6-deficient placentae, and reduced Ca2+ uptake of primary trophoblasts upon functional deletion of TRPV6. Trpv6-/- trophoblasts show a distinct calcium-dependent phenotype. Deep proteomic profiling of wt and Trpv6-/- primary trophoblasts using label-free quantitative mass spectrometry leads to the identification of 2778 proteins. Among those, a group of proteases, including high-temperature requirement A serine peptidase 1 (HTRA1) and different granzymes are more abundantly expressed in Trpv6-/- trophoblast lysates, whereas the extracellular matrix protein fibronectin and the fibronectin-domain-containing protein 3A (FND3A) were markedly reduced. Trpv6-/-placenta lysates contain a higher intrinsic proteolytic activity increasing fibronectin degradation. Our results show that the extracellular matrix formation of the placental labyrinth depends on TRPV6; its deletion in trophoblasts correlates with the increased expression of proteases controlling the extracellular matrix in the labyrinth during pregnancy.


Asunto(s)
Matriz Extracelular/metabolismo , Placenta/metabolismo , Canales Catiónicos TRPV/metabolismo , Transporte Biológico , Biomarcadores , Calcio/metabolismo , Movimiento Celular/genética , Supervivencia Celular/genética , Biología Computacional , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Embarazo , Proteolisis , Proteoma , Proteómica , Canales Catiónicos TRPV/genética
4.
J Struct Biol ; 199(2): 102-113, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28559167

RESUMEN

TMEM16A is a membrane protein forming a calcium-activated chloride channel. A homodimeric stoichiometry of the TMEM16 family of proteins has been reported but an important question is whether the protein resides always in a dimeric configuration in the plasma membrane or whether monomers of the protein are also present in its native state within in the intact plasma membrane. We have determined the stoichiometry of the human (h)TMEM16A within whole COS-7 cells in liquid. For the purpose of detecting TMEM16A subunits, single proteins were tagged by the streptavidin-binding peptide within extracellular loops accessible by streptavidin coated quantum dot (QD) nanoparticles. The labeled proteins were then imaged using correlative light microscopy and environmental scanning electron microscopy (ESEM) using scanning transmission electron microscopy (STEM) detection. The locations of 19,583 individual proteins were determined of which a statistical analysis using the pair correlation function revealed the presence of a dimeric conformation of the protein. The amounts of detected label pairs and single labels were compared between experiments in which the TMEM16A SBP-tag position was varied, and experiments in which tagged and non-tagged TMEM16A proteins were present. It followed that hTMEM16A resides in the plasma membrane as dimer only and is not present as monomer. This strategy may help to elucidate the stoichiometry of other membrane protein species within the context of the intact plasma membrane in future.


Asunto(s)
Anoctamina-1/análisis , Membrana Celular/química , Microscopía Electrónica de Transmisión de Rastreo/métodos , Multimerización de Proteína , Animales , Anoctamina-1/química , Células COS , Canales de Cloruro/análisis , Canales de Cloruro/química , Chlorocebus aethiops , Humanos , Subunidades de Proteína/análisis , Puntos Cuánticos , Coloración y Etiquetado/métodos , Estreptavidina
5.
Glia ; 65(9): 1535-1549, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28636132

RESUMEN

Following brain injury astrocytes change into a reactive state, proliferate and grow into the site of lesion, a process called astrogliosis, initiated and regulated by changes in cytoplasmic Ca2+ . Transient receptor potential canonical (TRPC) channels may contribute to Ca2+ influx but their presence and possible function in astrocytes is not known. By RT-PCR and RNA sequencing we identified transcripts of Trpc1, Trpc2, Trpc3, and Trpc4 in FACS-sorted glutamate aspartate transporter (GLAST)-positive cultured mouse cortical astrocytes and subcloned full-length Trpc1 and Trpc3 cDNAs from these cells. Ca2+ entry in cortical astrocytes depended on TRPC3 and was increased in the absence of Trpc1. After co-expression of Trpc1 and Trpc3 in HEK-293 cells both proteins co-immunoprecipitate and form functional heteromeric channels, with TRPC1 reducing TRPC3 activity. In vitro, lack of Trpc3 reduced astrocyte proliferation and migration whereas the TRPC3 gain-of-function moonwalker mutation and Trpc1 deficiency increased astrocyte migration. In vivo, astrogliosis and cortex edema following stab wound injury were reduced in Trpc3-/- but increased in Trpc1-/- mice. In summary, our results show a decisive contribution of TRPC3 to astrocyte Ca2+ signaling, which is even augmented in the absence of Trpc1, in particular following brain injury. Targeted therapies to reduce TRPC3 channel activity in astrocytes might therefore be beneficial in traumatic brain injury.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio/fisiología , Corteza Cerebral/lesiones , Gliosis/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Astrocitos/patología , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Gliosis/etiología , Gliosis/patología , Células HEK293 , Humanos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6 , Heridas Punzantes/metabolismo , Heridas Punzantes/patología
6.
J Biol Chem ; 289(14): 9766-80, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24563462

RESUMEN

TRPC4 proteins function as Ca(2+) conducting, non-selective cation channels in endothelial, smooth muscle, and neuronal cells. To further characterize the roles of TRPC4 in vivo, detailed information about the molecular composition of native channel complexes and their association with cellular signaling networks is needed. Therefore, a mouse brain cDNA library was searched for novel TRPC4-interacting proteins using a modified yeast two-hybrid assay. This screen identified Trans-activation Response RNA-binding protein 2 (Tarpb2), a protein that recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Tarbp2 was found to bind to the C terminus of TRPC4 and TRPC5 and to modulate agonist-dependent TRPC4-induced Ca(2+) entry. A stretch of basic residues within the Tarbp2 protein is required for these actions. Tarbp2 binding to and modulation of TRPC4 occurs in the presence of endogenously expressed Dicer but is no longer detectable when the Dicer cDNA is overexpressed. Dicer activity in crude cell lysates is increased in the presence of Ca(2+), most probably by Ca(2+)-dependent proteolytic activation of Dicer. Apparently, Tarbp2 binding to TRPC4 promotes changes of cytosolic Ca(2+) and, thereby, leads to a dynamic regulation of Dicer activity, essentially at low endogenous Dicer concentrations.


Asunto(s)
Calcio/metabolismo , MicroARNs/biosíntesis , Procesamiento Postranscripcional del ARN/fisiología , Proteínas de Unión al ARN/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Citosol/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Células HEK293 , Humanos , Ratones , MicroARNs/genética , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Canales Catiónicos TRPC/genética
7.
J Biol Chem ; 288(27): 19471-83, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23677990

RESUMEN

TRPC4 and TRPC5 proteins share 65% amino acid sequence identity and form Ca(2+)-permeable nonselective cation channels. They are activated by stimulation of receptors coupled to the phosphoinositide signaling cascade. Replacing a conserved glycine residue within the cytosolic S4-S5 linker of both proteins by a serine residue forces the channels into an open conformation. Expression of the TRPC4G503S and TRPC5G504S mutants causes cell death, which could be prevented by buffering the Ca(2+) of the culture medium. Current-voltage relationships of the TRPC4G503S and TRPC5G504S mutant ion channels resemble that of fully activated TRPC4 and TRPC5 wild-type channels, respectively. Modeling the structure of the transmembrane domains and the pore region (S4-S6) of TRPC4 predicts a conserved serine residue within the C-terminal sequence of the predicted S6 helix as a potential interaction site. Introduction of a second mutation (S623A) into TRPC4G503S suppressed the constitutive activation and partially rescued its function. These results indicate that the S4-S5 linker is a critical constituent of TRPC4/C5 channel gating and that disturbance of its sequence allows channel opening independent of any sensor domain.


Asunto(s)
Activación del Canal Iónico/fisiología , Canales Catiónicos TRPC/metabolismo , Sustitución de Aminoácidos , Animales , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Mutación Missense , Mapeo Peptídico , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Canales Catiónicos TRPC/genética
8.
J Biol Chem ; 288(23): 16629-16644, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23612980

RESUMEN

TRPV6 channels function as epithelial Ca(2+) entry pathways in the epididymis, prostate, and placenta. However, the identity of the endogenous TRPV6 protein relies on predicted gene coding regions and is only known to a certain level of approximation. We show that in vivo the TRPV6 protein has an extended N terminus. Translation initiates at a non-AUG codon, at ACG, which is decoded by methionine and which is upstream of the annotated AUG, which is not used for initiation. The in vitro properties of channels formed by the extended full-length TRPV6 proteins and the so-far annotated and smaller TRPV6 are similar, but the extended N terminus increases trafficking to the plasma membrane and represents an additional scaffold for channel assembly. The increased translation of the smaller TRPV6 cDNA version may overestimate the in vivo situation where translation efficiency may represent an additional mechanism to tightly control the TRPV6-mediated Ca(2+) entry to prevent deleterious Ca(2+) overload.


Asunto(s)
Canales de Calcio/biosíntesis , Membrana Celular/metabolismo , Codón Iniciador/metabolismo , Biosíntesis de Proteínas/fisiología , Canales Catiónicos TRPV/biosíntesis , Canales de Calcio/genética , Membrana Celular/genética , Codón Iniciador/genética , Células HEK293 , Humanos , Metionina , Transporte de Proteínas/fisiología , Canales Catiónicos TRPV/genética
9.
Handb Exp Pharmacol ; 222: 359-84, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24756713

RESUMEN

TRPV6 (former synonyms ECAC2, CaT1, CaT-like) displays several specific features which makes it unique among the members of the mammalian Trp gene family (1) TRPV6 (and its closest relative, TRPV5) are the only highly Ca(2+)-selective channels of the entire TRP superfamily (Peng et al. 1999; Wissenbach et al. 2001; Voets et al. 2004). (2) Translation of Trpv6 initiates at a non-AUG codon, at ACG, located upstream of the annotated AUG, which is not used for initiation (Fecher-Trost et al. 2013). The ACG codon is nevertheless decoded by methionine. Not only a very rare event in eukaryotic biology, the full-length TRPV6 protein existing in vivo comprises an amino terminus extended by 40 amino acid residues compared to the annotated truncated TRPV6 protein which has been used in most studies on TRPV6 channel activity so far. (In the following numbering occurs according to this full-length protein, with the numbers of the so far annotated truncated protein in brackets). (3) Only in humans a coupled polymorphism of Trpv6 exists causing three amino acid exchanges and resulting in an ancestral Trpv6 haplotype and a so-called derived Trpv6 haplotype (Wissenbach et al. 2001). The ancestral allele encodes the amino acid residues C197(157), M418(378) and M721(681) and the derived alleles R197(157), V418(378) and T721(681). The ancestral haplotype is found in all species, the derived Trpv6 haplotype has only been identified in humans, and its frequency increases with the distance to the African continent. Apparently the Trpv6 gene has been a strong target for selection in humans, and its derived variant is one of the few examples showing consistently differences to the orthologues genes of other primates (Akey et al. 2004, 2006; Stajich and Hahn 2005; Hughes et al. 2008). (4) The Trpv6 gene expression is significantly upregulated in several human malignancies including the most common cancers, prostate and breast cancer (Wissenbach et al. 2001; Zhuang et al. 2002; Fixemer et al. 2003; Bolanz et al. 2008). (5) Male mice lacking functional TRPV6 channels are hypo-/infertile making TRPV6 one of the very few channels essential for male fertility (Weissgerber et al. 2011, 2012).


Asunto(s)
Canales de Calcio/metabolismo , Canales Catiónicos TRPV/metabolismo , Secuencia de Aminoácidos , Animales , Canales de Calcio/química , Canales de Calcio/deficiencia , Canales de Calcio/efectos de los fármacos , Canales de Calcio/genética , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Potenciales de la Membrana , Moduladores del Transporte de Membrana/farmacología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Fenotipo , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/genética
10.
J Clin Invest ; 134(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38557489

RESUMEN

Regulated exocytosis is initiated by increased Ca2+ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca2+ directly released from acidic Ca2+ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca2+ regulator protein integral to the membrane of secretory granules that controlled Ca2+ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca2+ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca2+ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage.


Asunto(s)
Canales de Calcio , Calcio , Ratones , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Calcio/metabolismo , Páncreas/metabolismo , Exocitosis/fisiología , Vesículas Secretoras/genética
11.
J Biol Chem ; 287(22): 17930-41, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22427671

RESUMEN

Replacement of aspartate residue 541 by alanine (D541A) in the pore of TRPV6 channels in mice disrupts Ca(2+) absorption by the epididymal epithelium, resulting in abnormally high Ca(2+) concentrations in epididymal luminal fluid and in a dramatic but incomplete loss of sperm motility and fertilization capacity, raising the possibility of residual activity of channels formed by TRPV6(D541A) proteins (Weissgerber, P., Kriebs, U., Tsvilovskyy, V., Olausson, J., Kretz, O., Stoerger, C., Vennekens, R., Wissenbach, U., Middendorff, R., Flockerzi, V., and Freichel, M. (2011) Sci. Signal. 4, ra27). It is known from other cation channels that introducing pore mutations even if they largely affect their conductivity and permeability can evoke considerably different phenotypes compared with the deletion of the corresponding protein. Therefore, we generated TRPV6-deficient mice (Trpv6(-/-)) by deleting exons encoding transmembrane domains with the pore-forming region and the complete cytosolic C terminus harboring binding sites for TRPV6-associated proteins that regulate its activity and plasma membrane anchoring. Using this strategy, we aimed to determine whether the TRPV6(D541A) pore mutant still contributes to residual channel activity and/or channel-independent functions in vivo. Trpv6(-/-) males reveal severe defects in fertility and motility and viability of sperm and a significant increase in epididymal luminal Ca(2+) concentration that is mirrored by a lack of Ca(2+) uptake by the epididymal epithelium. Therewith, Trpv6 excision affects epididymal Ca(2+) handling and male fertility to the same extent as the introduction of the D541A pore mutation, arguing against residual functions of the TRPV6(D541A) pore mutant in epididymal epithelial cells.


Asunto(s)
Canales de Calcio/genética , Calcio/metabolismo , Epidídimo/metabolismo , Fertilidad/genética , Eliminación de Gen , Mutación , Canales Catiónicos TRPV/genética , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Masculino , Ratones , Ratones Noqueados , Motilidad Espermática/genética
12.
J Biol Chem ; 287(44): 36663-72, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22961981

RESUMEN

TRPM3 channels form ionotropic steroid receptors in the plasma membrane of pancreatic ß and dorsal root ganglion cells and link steroid hormone signaling to insulin release and pain perception, respectively. We identified and compared the function of a number of TRPM3 splice variants present in mouse, rat and human tissues. We found that variants lacking a region of 18 amino acid residues display neither Ca(2+) entry nor ionic currents when expressed alone. Hence, splicing removes a region that is indispensable for channel function, which is called the ICF region. TRPM3 variants devoid of this region (TRPM3ΔICF), are ubiquitously present in different tissues and cell types where their transcripts constitute up to 15% of the TRPM3 isoforms. The ICF region is conserved throughout the TRPM family, and its presence in TRPM8 proteins is also necessary for function. Within the ICF region, 10 amino acid residues form a domain essential for the formation of operative TRPM3 channels. TRPM3ΔICF variants showed reduced interaction with other TRPM3 isoforms, and their occurrence at the cell membrane was diminished. Correspondingly, coexpression of ΔICF proteins with functional TRPM3 subunits not only reduced the number of channels but also impaired TRPM3-mediated Ca(2+) entry. We conclude that TRPM3ΔICF variants are regulatory channel subunits fine-tuning TRPM3 channel activity.


Asunto(s)
Empalme Alternativo , Canales Catiónicos TRPM/genética , Secuencia de Aminoácidos , Animales , Señalización del Calcio , Secuencia Conservada , Exones , Células HEK293 , Humanos , Inmunoprecipitación , Potenciales de la Membrana , Ratones , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sitios de Empalme de ARN , Ratas , Homología de Secuencia de Aminoácido , Canales Catiónicos TRPM/química , Canales Catiónicos TRPM/metabolismo
13.
Cell Calcium ; 106: 102634, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35917684

RESUMEN

Amongst the superfamily of transient receptor potential (TRP) channels, TRPV5 and TRPV6 are specialized members that mediate Ca2+-selective transport across epithelial membranes. Intriguingly, fluorescent fusion proteins of TRPV5 or TRPV6 are hardly discernible within the plasma membrane of living cells. Instead, TRPV6 is mostly found in vesicular membrane compartments, indicating either a rapid degradation or cycling of channel-bearing vesicles between endomembrane compartments and the plasma membrane. In TRPV6-expressing cells, brefeldin A, a toxin that blocks the transit between the endoplasmic reticulum and the Golgi apparatus, caused a drop in [Ca2+]i with a half time in the range of 0.5-1 h. Upon wash-out of the toxin, the [Ca2+]i rose to a steady-state level within 2-3 h. Consistently, the synchronized forward trafficking of TRPV6VL-eGFP after brefeldin A wash-out led to a visible accumulation of the protein within the plasma membrane, as shown by confocal and total internal reflection microscopy. Analysis of the internalization route and differentiation of vesicle populations provided evidence for a clathrin-dependent internalization pathway. Most TRPV6VL-bearing vesicles co-stained with Rab5a, a marker protein for early endosomes. Fewer vesicles were co-localized with Rab7a (late endosomes) or with Rab11 (recycling endosomes). From these data, we propose that the lack of plasma membrane visibility of the channel results from a rapid internalization, which in addition to transcriptional regulation, adds a layer of functional channel regulation to modulate transepithelial Ca2+ transport.


Asunto(s)
Calcio , Canales Catiónicos TRPV , Brefeldino A/metabolismo , Brefeldino A/farmacología , Calcio/metabolismo , Membrana Celular/metabolismo , Aparato de Golgi/metabolismo , Canales Catiónicos TRPV/metabolismo
14.
Ann Anat ; 238: 151753, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33964462

RESUMEN

It is well known that not all biological findings derived from animals can be directly applied to humans. The TRPV6 protein may serve as an example which highlights these inter-species differences as an example of parallel evolutionary pathways. TRPV6 (and TRPV5) belong to a family of ion channels from the transient receptor potential group but are selectively permeable for Ca2+, in contrast to other members of the family. Sequences with recognizable similarity to TRPV6 can already be found in archaebacteria. These ancient sequences show clear similarity to the ion-conducting pore of TRPV6. Over the course of evolution, the duplication of the TRPV6 gene gave rise to TRPV5. Duplications of the complete genome as well as subsequent loss of genetic material have led to a variety of different TRPV5/6 combinations. In addition, there is an N-terminal extension of the protein in placental animals. This extension causes translation of TRPV6 to be initiated from an ACG codon. Inactivation of one TRPV6 allele can be correlated with alcohol-independent pancreatitis in humans while inactivation of both alleles leads to skeletal dysplasia of newborn babies. The latter effect is not observed in mice, implying that the effects due to perturbations in TRPV6 levels are much more pronounced in humans.


Asunto(s)
Canales de Calcio , Canales Catiónicos TRPV , Animales , Calcio/metabolismo , Femenino , Ratones , Placenta , Embarazo , Canales Catiónicos TRPV/genética
15.
Biochim Biophys Acta Gen Subj ; 1865(6): 129873, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33610740

RESUMEN

The TRP-family of ion channels consists of 27 members in humans. Most TRP channels are non- selective cation channels with the exception of TRPV5 and TRPV6 which exhibit a high permeability for Ca2+ ions. A functional channel is formed by 4 identical subunits [1]. A growing number of mutations are present in human TRPV6 genes which alter channel function and can lead to elevated blood levels of the parathyroid hormone accompanied by transient hyperparathyroidism. Recent publications suggest that TRPV6 mutations could also trigger non-alcoholic chronic pancreatitis. This review summarises the consequences of these mutations within the TRPV6 gene.


Asunto(s)
Canales de Calcio/genética , Calcio/metabolismo , Canalopatías/patología , Mutación , Canales Catiónicos TRPV/genética , Canales de Calcio/metabolismo , Canalopatías/etiología , Humanos , Canales Catiónicos TRPV/metabolismo
16.
J Biol Chem ; 284(49): 34423-32, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19815560

RESUMEN

TRPC5 forms non-selective cation channels. Here we studied the role of internal Ca(2+) in the activation of murine TRPC5 heterologously expressed in human embryonic kidney cells. Cell dialysis with various Ca(2+) concentrations (Ca(2+)(i)) revealed a dose-dependent activation of TRPC5 channels by internal Ca(2+) with EC(50) of 635.1 and 358.2 nm at negative and positive membrane potentials, respectively. Stepwise increases of Ca(2+)(i) induced by photolysis of caged Ca(2+) showed that the Ca(2+) activation of TRPC5 channels follows a rapid exponential time course with a time constant of 8.6 +/- 0.2 ms at Ca(2+)(i) below 10 microM, suggesting that the action of internal Ca(2+) is a primary mechanism in the activation of TRPC5 channels. A second slow activation phase with a time to peak of 1.4 +/- 0.1 s was also observed at Ca(2+)(i) above 10 microM. In support of a Ca(2+)-activation mechanism, the thapsigargin-induced release of Ca(2+) from internal stores activated TRPC5 channels transiently, and the subsequent Ca(2+) entry produced a sustained TRPC5 activation, which in turn supported a long-lasting membrane depolarization. By co-expressing STIM1 plus ORAI1 or the alpha(1)C and beta(2) subunits of L-type Ca(2+) channels, we found that Ca(2+) entry through either calcium-release-activated-calcium or voltage-dependent Ca(2+) channels is sufficient for TRPC5 channel activation. The Ca(2+) entry activated TRPC5 channels under buffering of internal Ca(2+) with EGTA but not with BAPTA. Our data support the hypothesis that TRPC5 forms Ca(2+)-activated cation channels that are functionally coupled to Ca(2+)-selective ion channels through local Ca(2+) increases beneath the plasma membrane.


Asunto(s)
Canales Iónicos/química , Canales Catiónicos TRPC/metabolismo , Animales , Calcio/química , Calcio/metabolismo , Línea Celular , Membrana Celular/metabolismo , Quelantes/farmacología , Humanos , Iones , Potenciales de la Membrana , Ratones , Modelos Biológicos , Canales Catiónicos TRPC/fisiología , Tapsigargina/química , Factores de Tiempo
17.
Nature ; 430(7001): 748-54, 2004 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-15306801

RESUMEN

The mammalian sensory system is capable of discriminating thermal stimuli ranging from noxious cold to noxious heat. Principal temperature sensors belong to the TRP cation channel family, but the mechanisms underlying the marked temperature sensitivity of opening and closing ('gating') of these channels are unknown. Here we show that temperature sensing is tightly linked to voltage-dependent gating in the cold-sensitive channel TRPM8 and the heat-sensitive channel TRPV1. Both channels are activated upon depolarization, and changes in temperature result in graded shifts of their voltage-dependent activation curves. The chemical agonists menthol (TRPM8) and capsaicin (TRPV1) function as gating modifiers, shifting activation curves towards physiological membrane potentials. Kinetic analysis of gating at different temperatures indicates that temperature sensitivity in TRPM8 and TRPV1 arises from a tenfold difference in the activation energies associated with voltage-dependent opening and closing. Our results suggest a simple unifying principle that explains both cold and heat sensitivity in TRP channels.


Asunto(s)
Frío , Calor , Activación del Canal Iónico , Canales Iónicos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Droga/metabolismo , Capsaicina/farmacología , Línea Celular , Conductividad Eléctrica , Humanos , Activación del Canal Iónico/efectos de los fármacos , Canales Iónicos/agonistas , Canales Iónicos/genética , Ligandos , Potenciales de la Membrana/efectos de los fármacos , Mentol/farmacología , Modelos Biológicos , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/genética , Técnicas de Placa-Clamp , Receptores de Droga/agonistas , Receptores de Droga/genética , Canales Catiónicos TRPM
18.
Biochem Pharmacol ; 177: 113936, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32224140

RESUMEN

The TRPM8 cation channel can be activated by the cooling compound icilin. Recently, we showed that stimulation of TRPM8 channels induces a signaling cascade leading to the activation of the transcription factor AP-1. Additionally, expression of the AP-1 constituent c-Fos has been shown to be induced following TRPM8 stimulation. c-Fos is frequently used as a marker for neuronal activity. Here, we have analyzed the mechanism connecting TRPM8 stimulation and c-Fos expression. Furthermore, we analyzed the expression of the neuronal activity-responsive transcription factor Egr-1 following TRPM8 activation. The results show that icilin-induced stimulation of TRPM8 channels increased c-Fos promoter activity and induced c-Fos expression. Moreover, icilin stimulation increased Egr-1 promoter activity and induced the expression of Egr-1. Pharmacological inhibition of TRPM8 blocked the icilin-induced expression of Egr-1 and c-Fos. An influx of Ca2+ ions into the cells via TRPM8 was necessary to stimulate Egr-1 and c-Fos expression following icilin treatment. Genetic experiments revealed that serum response elements within the Egr-1 and c-Fos promoters are crucial to couple TRPM8 stimulation with enhanced transcription of both the Egr-1 and c-Fos genes. These data were corroborated by experiments showing that TRPM8 stimulation increased the transcriptional activation potential of Elk-1, a SRE binding protein. c-Fos is important for neuronal excitability and survival. Egr-1 plays an important role in synaptic plasticity, consolidation and reconsolidation of long-term memory. Elk-1 may preserve neurons against toxic insults but may also induce depressive behaviour. The fact that TRPM8 stimulation activates the transcription factors c-Fos, Egr-1, and Elk-1 connects TRPM8 signaling with maintaining important brain functions.


Asunto(s)
Calcio/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Genes fos , Iones/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Pirimidinonas/farmacología , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPM/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Canales de Calcio/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Células HEK293 , Humanos , Regiones Promotoras Genéticas , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Proteína Elk-1 con Dominio ets/genética
19.
Cell Calcium ; 92: 102302, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33069057

RESUMEN

TRPV6 is a calcium selective TRP channel and is expressed in many species. TRPV6 transcripts are abundantly expressed in few tissues but strangely channel properties are only accessible to electrophysiological recordings after overexpression whereas in native tissue functional channel currents seem not to be detectable. Another exceptional property of human and mouse TRPV6 proteins is that the initiation of translation starts from a non-canonical ACG triplet which is translated as methionine. This triplet is located 120 bp upstream of the first in-frame AUG codon of the human/mouse TRPV6 mRNA. In contrast, the TRPV6 gene of bats is initiated from an AUG triplet at the corresponding position of the human ACG. On the basis of these structural nucleotide differences between human and bats we studied the role of the absolute N-Terminus for the regulation of translation by developing chimera and mutants of human/bat TRPV6 channels. The human sequence which is located downstream of the initiation codon slows down ribosomal scanning in 3' direction. We suggest that the mechanism involves most likely the deceleration of ribosome scanning by stem-loop formation and the use of the common initiator tRNA, tRNAiMet, which is placed onto the inappropriate ACG codon resulting in low protein synthesis. The reduced translation efficiency is important to protect TRPV6 expressing cells from toxic calcium overload. The regulation of the TRPV6 translation in bats may be an adaptation to low calcium amounts present in the natural nutrition. In addition, we show that also the GFP protein can be controlled using the translational mechanism of human TRPV6.


Asunto(s)
Quirópteros/fisiología , Activación del Canal Iónico , Canales Catiónicos TRPV/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Calcio/metabolismo , Células HEK293 , Humanos , Mutación/genética , Filogenia , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/genética
20.
BMC Cancer ; 9: 380, 2009 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-19857260

RESUMEN

BACKGROUND: The transient receptor potential, subfamily V, member 6 (TRPV6) is a Ca(2+) selective cation channel. Several studies have shown that TRPV6 transcripts are expressed in locally advanced prostatic adenocarcinoma, metastatic and androgen-insensitive prostatic lesions but are undetectable in healthy prostate tissue and benign prostatic hyperplasia. Two allelic variants of the human trpv6 gene have been identified which are transcribed into two independent mRNAs, TRPV6a and TRPV6b. We now asked, whether the trpv6a allele is correlated with the onset of prostate cancer, with the Gleason score and the tumour stage. METHODS: Genomic DNA of prostate cancer patients and control individuals was isolated from resections of prostatic adenocarcinomas and salivary fluid respectively. Genotyping of SNPs of the TRPV6 gene was performed by restriction length polymorphism or by sequencing analysis. RNA used for RT-PCR was isolated from prostate tissue. Data sets were analyzed by Chi-Square test. RESULTS: We first characterized in detail the five polymorphisms present in the protein coding exons of the trpv6 gene and show that these polymorphisms are coupled and are underlying the TRPV6a and the TRPV6b variants. Next we analysed the frequencies of the two TRPV6 alleles using genomic DNA from saliva samples of 169 healthy individuals. The homozygous TRPV6b genotype predominated with 86%, whereas no homozygous TRPV6a carriers could be identified. The International HapMap Project identified a similar frequency for an Utah based population whereas in an African population the a-genotype prevailed. The incidence of prostate cancer is several times higher in African populations than in non-African and we then investigated the TRPV6a/b frequencies in 141 samples of prostatic adenocarcinoma. The TRPV6b allele was found in 87% of the samples without correlation with Gleason score and tumour stage. CONCLUSION: Our results show that the frequencies of trpv6 alleles in healthy control individuals and prostate cancer patients are not significantly different. Although expression of trpv6 transcripts correlates with aggressive potential of prostate cancer, the TRPV6 genotype does not correlate with the onset of prostate cancer, with the Gleason score and the tumour stage.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Canales de Calcio/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Canales Catiónicos TRPV/genética , Anciano , Alelos , Estudios de Casos y Controles , Estudios de Cohortes , Progresión de la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo Genético , Grupos Raciales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA