Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pflugers Arch ; 476(6): 923-937, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38627262

RESUMEN

Fast growing solid tumors are frequently surrounded by an acidic microenvironment. Tumor cells employ a variety of mechanisms to survive and proliferate under these harsh conditions. In that regard, acid-sensitive membrane receptors constitute a particularly interesting target, since they can affect cellular functions through ion flow and second messenger cascades. Our knowledge of these processes remains sparse, however, especially regarding medulloblastoma, the most common pediatric CNS malignancy. In this study, using RT-qPCR, whole-cell patch clamp, and Ca2+-imaging, we uncovered several ion channels and a G protein-coupled receptor, which were regulated directly or indirectly by low extracellular pH in DAOY and UW228 medulloblastoma cells. Acidification directly activated acid-sensing ion channel 1a (ASIC1a), the proton-activated Cl- channel (PAC, ASOR, or TMEM206), and the proton-activated G protein-coupled receptor OGR1. The resulting Ca2+ signal secondarily activated the large conductance calcium-activated potassium channel (BKCa). Our analyses uncover a complex relationship of these transmembrane proteins in DAOY cells that resulted in cell volume changes and induced cell death under strongly acidic conditions. Collectively, our results suggest that these ion channels in concert with OGR1 may shape the growth and evolution of medulloblastoma cells in their acidic microenvironment.


Asunto(s)
Canales Iónicos Sensibles al Ácido , Meduloblastoma , Receptores Acoplados a Proteínas G , Humanos , Canales Iónicos Sensibles al Ácido/metabolismo , Canales Iónicos Sensibles al Ácido/genética , Meduloblastoma/metabolismo , Meduloblastoma/patología , Línea Celular Tumoral , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Concentración de Iones de Hidrógeno , Tamaño de la Célula , Muerte Celular , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Calcio/metabolismo , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología
2.
J Pineal Res ; 76(1): e12919, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37794846

RESUMEN

Besides its role in the circadian rhythm, the pineal gland hormone melatonin (MLT) also possesses antiepileptogenic, antineoplastic, and cardioprotective properties, among others. The dosages necessary to elicit beneficial effects in these diseases often far surpass physiological concentrations. Although even high doses of MLT are considered to be largely harmless to humans, the possible side effects of pharmacological concentrations are so far not well investigated. In the present study, we report that pharmacological doses of MLT (3 mM) strongly altered the electrophysiological characteristics of cultured primary mouse cerebellar granule cells (CGCs). Using whole-cell patch clamp and ratiometric Ca2+ imaging, we observed that pharmacological concentrations of MLT inhibited several types of voltage-gated Na+ , K+ , and Ca2+ channels in CGCs independently of known MLT-receptors, altering the character and pattern of elicited action potentials (APs) significantly, quickly and reversibly. Specifically, MLT reduced AP frequency, afterhyperpolarization, and rheobase, whereas AP amplitude and threshold potential remained unchanged. The altered biophysical profile of the cells could constitute a possible mechanism underlying the proposed beneficial effects of MLT in brain-related disorders, such as epilepsy. On the other hand, it suggests potential adverse effects of pharmacological MLT concentrations on neurons, which should be considered when using MLT as a pharmacological compound.


Asunto(s)
Canales de Calcio , Melatonina , Humanos , Ratones , Animales , Canales de Calcio/farmacología , Canales de Calcio/fisiología , Melatonina/farmacología , Sodio/farmacología , Potasio/farmacología , Neuronas/metabolismo , Calcio/metabolismo
3.
Pflugers Arch ; 475(9): 1073-1087, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37474775

RESUMEN

Acid-sensing ion channels (ASICs) are Na+ channels that are almost ubiquitously expressed in neurons of the brain. Functional ASIC1a is also expressed in glioblastoma stem cells, where it might sense the acidic tumor microenvironment. Prolonged acidosis induces cell death in neurons and reduces tumor sphere formation in glioblastoma via activation of ASIC1a. It is currently unknown whether ASICs are expressed and involved in acid-induced cell death in other types of brain tumors. In this study, we investigated ASICs in medulloblastoma, using two established cell lines, DAOY and UW228, as in vitro models. In addition, we characterized ASICs in the most numerous neuron of the brain, the cerebellar granule cell, which shares the progenitor cell with some forms of medulloblastoma. We report compelling evidence using RT-qPCR, western blot and whole-cell patch clamp that DAOY and cerebellar granule cells, but not UW228 cells, functionally express homomeric ASIC1a. Additionally, Ca2+-imaging revealed that extracellular acidification elevated intracellular Ca2+-levels in DAOY cells independently of ASICs. Finally, we show that overexpression of RIPK3, a key component of the necroptosis pathway, renders DAOY cells susceptible to acid-induced cell death via activation of ASIC1a. Our data support the idea that ASIC1a is an important acid sensor in brain tumors and that its activation has potential to induce cell death in tumor cells.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Cerebelosas , Glioblastoma , Meduloblastoma , Humanos , Canales Iónicos Sensibles al Ácido/metabolismo , Meduloblastoma/metabolismo , Glioblastoma/metabolismo , Neuronas/metabolismo , Línea Celular , Neoplasias Encefálicas/metabolismo , Cerebelo , Neoplasias Cerebelosas/metabolismo , Microambiente Tumoral
4.
ACS Chem Neurosci ; 14(14): 2487-2498, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37379568

RESUMEN

Acid-sensing ion channels (ASICs) are proton-gated ion channels that contribute to pain perception and neurotransmission. Being involved in sensing inflammation and ischemia, ASIC1a and ASIC3 are promising drug targets. Polyphenol tannic acid (TA) as well as green tea can interact with a variety of ion channels, but their effect on ASICs remains unknown. In addition, it is unknown whether they interact with ion channels via a common mechanism. Here, we show that TA is a potent modulator of ASICs. TA inhibited the transient current of rat ASIC3 expressed in HEK cells with an apparent IC50 of 2.2 ± 0.6 µM; it potentiated the sustained current and induced a slowly declining decay current. In addition, it produced an acidic shift of the pH-dependent activation of ASIC3 and inhibited the window current at pH 7.0. Moreover, TA inhibited the transient current of ASIC1a, ASIC1b, and ASIC2a. Pentagalloylglucose that is chemically identical to the central part of TA and a green tea extract both had effects on ASIC3 comparable to TA. TA and green tea inhibited inward currents generated by gramicidin channels, indicating interaction with the membrane. These results show that TA, pentagalloylglucose, and green tea modulate ASICs and identify alteration of the membrane as the potential common mechanism of this modulation. These properties will limit clinical application of these molecules.


Asunto(s)
Canales Iónicos Sensibles al Ácido , , Ratas , Animales , Taninos Hidrolizables , Concentración de Iones de Hidrógeno
5.
Pflugers Arch ; 475(3): 405-416, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36522586

RESUMEN

The microenvironment of proliferative and aggressive tumours, such as the brain tumour glioblastoma multiforme (GBM), is often acidic, hypoxic, and nutrient deficient. Acid-sensing ion channels (ASICs) are proton-sensitive Na+ channels that have been proposed to play a role in pH sensing and in modulation of cancer cell migration. We previously reported that primary glioblastoma stem cells (GSCs), which grow as multicellular tumour spheroids, express functional ASIC1a and ASIC3, whereas ASIC2a is downregulated in GSCs. Using a 2.5D migration assay, here we report that acidic pH dramatically increased migration of GSCs of the pro-neural subtype. Pharmacological blockade as well as CRISPR-Cas9-mediated gene knock-out of ASIC1a or stable overexpression of ASIC2a, however, revealed that neither ASIC1a nor ASIC3, nor downregulation of ASIC2a, mediated the aggressive migration at acidic pH. Therefore, we tested the role of two other proteins previously implicated in cancer cell migration: the Ca2+-activated K+ channel KCa3.1 (KCNN4) and phosphoinositide 3-kinase (PI3K). While pharmacological blockade of KCa3.1 did also not affect migration, blockade of PI3K decreased migration at acidic pH to control levels. In summary, our study reveals a strongly enhanced migration of GSCs at acidic pH in vitro and identifies PI3K as an important mediator of this effect.


Asunto(s)
Glioblastoma , Humanos , Canales Iónicos Sensibles al Ácido/genética , Canales Iónicos Sensibles al Ácido/metabolismo , Concentración de Iones de Hidrógeno , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Microambiente Tumoral , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo
6.
Cell Death Dis ; 13(8): 702, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-35961983

RESUMEN

Eliciting regulated cell death, like necroptosis, is a potential cancer treatment. However, pathways eliciting necroptosis are poorly understood. It has been reported that prolonged activation of acid-sensing ion channel 1a (ASIC1a) induces necroptosis in mouse neurons. Glioblastoma stem cells (GSCs) also express functional ASIC1a, but whether prolonged activation of ASIC1a induces necroptosis in GSCs is unknown. Here we used a tumorsphere formation assay to show that slight acidosis (pH 6.6) induces necrotic cell death in a manner that was sensitive to the necroptosis inhibitor Nec-1 and to the ASIC1a antagonist PcTx1. In addition, genetic knockout of ASIC1a rendered GSCs resistant to acid-induced reduction in tumorsphere formation, while the ASIC1 agonist MitTx1 reduced tumorsphere formation also at neutral pH. Finally, a 20 amino acid fragment of the ASIC1 C-terminus, thought to interact with the necroptosis kinase RIPK1, was sufficient to reduce the formation of tumorspheres. Meanwhile, the genetic knockout of MLKL, the executive protein in the necroptosis cascade, did not prevent a reduction in tumor sphere formation, suggesting that ASIC1a induced an alternative cell death pathway. These findings demonstrate that ASIC1a is a death receptor on GSCs that induces cell death during prolonged acidosis. We propose that this pathway shapes the evolution of a tumor in its acidic microenvironment and that pharmacological activation of ASIC1a might be a potential new strategy in tumor therapy.


Asunto(s)
Acidosis , Glioblastoma , Canales Iónicos Sensibles al Ácido/genética , Canales Iónicos Sensibles al Ácido/metabolismo , Acidosis/metabolismo , Animales , Glioblastoma/genética , Glioblastoma/metabolismo , Ratones , Neuronas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Células Madre/metabolismo , Microambiente Tumoral
7.
Front Cell Neurosci ; 15: 668008, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33986647

RESUMEN

Acid-sensing ion channels (ASICs) are proton-gated Na+ channels. They contribute to synaptic transmission, neuronal differentiation and neurodegeneration. ASICs have been mainly characterized in neurons from mice or rats and our knowledge of their properties in human neurons is scarce. Here, we functionally characterized ASICs in differentiating LUHMES cells, a human mesencephalic cell line with characteristics of dopaminergic neurons. We find that LUHMES cells express functional ASICs, predominantly homomeric ASIC1a. Expression starts early during differentiation with a striking surge in current amplitude at days 4-6 of differentiation, a time point where-based on published data-LUHMES cells start expressing synaptic markers. Peak ASIC expression therefore coincides with a critical period of LUHMES cell differentiation. It was associated with increased excitability, but not paralleled by an increase in ASIC1 mRNA or protein. In differentiating as well as in terminally differentiated LUHMES cells, ASIC activation by slight acidification elicited large currents, action potentials and a rise in cytosolic Ca2+. Applying the ASIC pore blocker diminazene during differentiation reduced the length of neurites, consistent with the hypothesis that ASICs play a critical role in LUHMES cell differentiation. In summary, our study establishes LUHMES cells as a valuable model to study the role of ASICs for neuronal differentiation and potentially also cell death in a human cell line.

8.
BMC Biol ; 18(1): 143, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33059680

RESUMEN

BACKGROUND: ADP-ribosylation is a ubiquitous post-translational modification that involves both mono- and poly-ADP-ribosylation. ARTD10, also known as PARP10, mediates mono-ADP-ribosylation (MARylation) of substrate proteins. A previous screen identified protein kinase C delta (PKCδ) as a potential ARTD10 substrate, among several other kinases. The voltage-gated K+ channel Kv1.1 constitutes one of the dominant Kv channels in neurons of the central nervous system and the inactivation properties of Kv1.1 are modulated by PKC. In this study, we addressed the role of ARTD10-PKCδ as a regulator of Kv1.1. RESULTS: We found that ARTD10 inhibited PKCδ, which increased Kv1.1 current amplitude and the proportion of the inactivating current component in HeLa cells, indicating that ARTD10 regulates Kv1.1 in living cells. An inhibitor of ARTD10, OUL35, significantly decreased peak amplitude together with the proportion of the inactivating current component of Kv1.1-containing channels in primary hippocampal neurons, demonstrating that the ARTD10-PKCδ signaling cascade regulates native Kv1.1. Moreover, we show that the pharmacological blockade of ARTD10 increases excitability of hippocampal neurons. CONCLUSIONS: Our results, for the first time, suggest that MARylation by ARTD10 controls neuronal excitability.


Asunto(s)
Canal de Potasio Kv.1.1/genética , Poli(ADP-Ribosa) Polimerasas/genética , Proteína Quinasa C-delta/genética , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Animales , Células HEK293 , Células HeLa , Humanos , Canal de Potasio Kv.1.1/metabolismo , Ratones , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteínas Proto-Oncogénicas/metabolismo
9.
Sci Rep ; 8(1): 8764, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29884813

RESUMEN

The Kryptopterus bicirrhis (glass catfish) is known to respond to electromagnetic fields (EMF). Here we tested its avoidance behavior in response to static and alternating magnetic fields stimulation. Using expression cloning we identified an electromagnetic perceptive gene (EPG) from the K. bicirrhis encoding a protein that responds to EMF. This EPG gene was cloned and expressed in mammalian cells, neuronal cultures and in rat's brain. Immunohistochemistry showed that the expression of EPG is confined to the mammalian cell membrane. Calcium imaging in mammalian cells and cultured neurons expressing EPG demonstrated that remote activation by EMF significantly increases intracellular calcium concentrations, indicative of cellular excitability. Moreover, wireless magnetic activation of EPG in rat motor cortex induced motor evoked responses of the contralateral forelimb in vivo. Here we report on the development of a new technology for remote, non-invasive modulation of cell function.


Asunto(s)
Reacción de Prevención , Campos Electromagnéticos , Peces/fisiología , Animales , Calcio/metabolismo , Células Cultivadas , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Peces/genética , Células HEK293 , Humanos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Tecnología Inalámbrica
10.
Sci Rep ; 7(1): 13674, 2017 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-29057936

RESUMEN

Acidic microenvironment is commonly observed in tumour tissues, including glioblastoma (GBM), the most aggressive and lethal brain tumour in adults. Acid sensing ion channels (ASICs) are neuronal voltage-insensitive sodium channels, which are sensors of extracellular protons. Here we studied and functionally characterized ASICs in two primary glioblastoma stem cell lines as cell culture models. We detected transcripts of the ACCN2 and ACCN3 genes, coding for ASIC1 and ASIC3, respectively, but not transcripts of ACCN1 (coding for ASIC2). Available microarray data confirmed that ACCN1 is downregulated in glioma. Western blotting confirmed expression of ASIC1 and ASIC3, the most proton-sensitive ASICs, in both GBM cell lines. We characterized ASICs functionally using whole-cell patch clamp and detected different types of acid-sensitive currents. Some of these currents had kinetics typical for ASICs and were sensitive to specific toxin inhibitors of ASIC1a or ASIC3, demonstrating that the GBM cell lines express functional ASIC1a and ASIC3 that may enable GBM cells to sensitively detect extracellular pH in a tumour tissue. Microarray data revealed that expression of ACCN2 and ACCN3 is associated with improved survival of patients suffering from gliomas, suggesting that preserved susceptibility to extracellular pH may impair tumour growth.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Glioblastoma/metabolismo , Antígeno AC133/metabolismo , Biomarcadores de Tumor/metabolismo , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Línea Celular Tumoral , Espacio Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/mortalidad , Humanos , Concentración de Iones de Hidrógeno , Potenciales de la Membrana/fisiología , Células Madre Neoplásicas/metabolismo , ARN Mensajero/metabolismo
11.
Cell Death Differ ; 24(10): 1655-1671, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28622300

RESUMEN

Amyotrophic lateral sclerosis (ALS) is characterized by the selective degeneration of motor neurons (MNs) and their target muscles. Misfolded proteins which often form intracellular aggregates are a pathological hallmark of ALS. Disruption of the functional interplay between protein degradation (ubiquitin proteasome system and autophagy) and RNA-binding protein homeostasis has recently been suggested as an integrated model that merges several ALS-associated proteins into a common pathophysiological pathway. The E102Q mutation in one such candidate gene, the endoplasmic reticulum (ER) chaperone Sigma receptor-1 (SigR1), has been reported to cause juvenile ALS. Although loss of SigR1 protein contributes to neurodegeneration in several ways, the molecular mechanisms underlying E102Q-SigR1-mediated neurodegeneration are still unclear. In the present study, we showed that the E102Q-SigR1 protein rapidly aggregates and accumulates in the ER and associated compartments in transfected cells, leading to structural alterations of the ER, nuclear envelope and mitochondria and to subsequent defects in proteasomal degradation and calcium homeostasis. ER defects and proteotoxic stress generated by E102Q-SigR1 aggregates further induce autophagy impairment, accumulation of stress granules and cytoplasmic aggregation of the ALS-linked RNA-binding proteins (RBPs) matrin-3, FUS, and TDP-43. Similar ultrastructural abnormalities as well as altered protein degradation and misregulated RBP homeostasis were observed in primary lymphoblastoid cells (PLCs) derived from E102Q-SigR1 fALS patients. Consistent with these findings, lumbar α-MNs of both sALS as well as fALS patients showed cytoplasmic matrin-3 aggregates which were not co-localized with pTDP-43 aggregates. Taken together, our results support the notion that E102Q-SigR1-mediated ALS pathogenesis comprises a synergistic mechanism of both toxic gain and loss of function involving a vicious circle of altered ER function, impaired protein homeostasis and defective RBPs.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Estrés del Retículo Endoplásmico/genética , Homeostasis/genética , Mutación/genética , Proteínas de Unión al ARN/metabolismo , Receptores sigma/genética , Animales , Retículo Endoplásmico/metabolismo , Humanos , Ratones , Neuronas Motoras/metabolismo , ARN/metabolismo , Receptor Sigma-1
12.
Proc Natl Acad Sci U S A ; 114(17): E3507-E3515, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396446

RESUMEN

Acid-sensing ion channels (ASICs) are proton-gated Na+ channels that are expressed throughout the nervous system. ASICs have been implicated in several neuronal disorders, like ischemic stroke, neuronal inflammation, and pathological pain. Several toxins from venomous animals have been identified that target ASICs with high specificity and potency. These toxins are extremely useful in providing protein pharmacophores and to characterize function and structure of ASICs. Marine cone snails contain a high diversity of toxins in their venom such as conotoxins, which are short polypeptides stabilized by disulfide bonds, and conopeptides, which have no or only one disulfide bond. Whereas conotoxins selectively target specific neuronal proteins, mainly ion channels, the targets of conopeptides are less well known. Here, we perform an in vitro screen of venoms from 18 cone snail species to identify toxins targeting ASICs. We identified a small conopeptide of only four amino acids from the venom of Conus textile that strongly potentiated currents of ASIC3, which has a specific role in the pain pathway. This peptide, RPRFamide, belongs to the subgroup of cono-RFamides. Electrophysiological characterization of isolated dorsal root ganglion (DRG) neurons revealed that RPRFamide increases their excitability. Moreover, injection of the peptide into the gastrocnemius muscle strongly enhanced acid-induced muscle pain in mice that was abolished by genetic inactivation of ASIC3. In summary, we identified a conopeptide that targets the nociceptor-specific ion channel ASIC3.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Conotoxinas/química , Conotoxinas/toxicidad , Caracol Conus/química , Ganglios Espinales/metabolismo , Músculo Esquelético/metabolismo , Mialgia/metabolismo , Neuronas/metabolismo , Animales , Ganglios Espinales/patología , Ratones , Músculo Esquelético/fisiología , Mialgia/inducido químicamente , Mialgia/patología , Neuronas/patología , Xenopus laevis
13.
Cell Signal ; 26(12): 2826-33, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25220380

RESUMEN

Eight paralogue members form the family of transmembrane channel-like (TMC) proteins that share considerable sequence homology to anoctamin 1 (Ano1, TMEM16A). Ano1 is a Ca(2+) activated Cl(-) channel that is related to head and neck cancer, often caused by human papilloma virus (HPV) infection. Mutations in TMC 6 and 8 (EVER1, EVER2) cause epidermodysplasia verruciformis. This rare skin disease is characterized by abnormal susceptibility to HPV infection and cancer. We found that in contrast to Ano1 the common paralogues TMC4-TMC8 did not produce Ca(2+) activated Cl(-) currents when expressed in HEK293 cells. On the contrary, TMC8 was found to be localized in the endoplasmic reticulum (ER), where it inhibited receptor mediated Ca(2+) release, activation of Ano1 and volume regulated LRRC8-related Cl(-) currents. Zn(2+) is co-released from the ER together with Ca(2+) and thereby further augments Ca(2+) store release. Because TMC8 is required to lower cytosolic Zn(2+) concentrations by the Zn(2+) transporter ZnT-1, we hypothesize that HPV infections and cancer caused by mutations in TMC8 are related to upregulated Zn(2+)/Ca(2+) signaling and activation of Ano1.


Asunto(s)
Calcio/farmacología , Canales de Cloruro/metabolismo , Cloruros/metabolismo , Espacio Intracelular/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Transducción de Señal/efectos de los fármacos , Zinc/farmacología , Adenosina Trifosfato/farmacología , Animales , Anoctamina-1 , Señalización del Calcio/efectos de los fármacos , Canales de Cloruro/antagonistas & inhibidores , Conductividad Eléctrica , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Ionomicina/farmacología , Ionóforos/farmacología , Modelos Biológicos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Ratas
14.
Biochem Biophys Res Commun ; 451(2): 258-62, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25094048

RESUMEN

Anoctamin 5 (Ano5) belongs to the anoctamin gene family and acts as a calcium-activated chloride channel (CaCC). A mutation in the Ano5 gene causes limb-girdle muscular dystrophy (LGMD) type 2L, the third most common LGMD in Northern and Central Europe. Defective sarcolemmal membrane repair has been reported in patients carrying this Ano5 mutant. It has also been noted that LGMD patients often suffer from nonspecific pharyngoesophageal motility disorders. One study reported that 8/19 patients carrying Ano5 nutations suffered from dysphagia, including the feeling that solid food items become lodged in the upper portion of the esophagus. Ano5 is widely distributed in bone, skeletal muscle, cardiac muscle, brain, heart, kidney and lung tissue, but no report has examined its expression in the gastrointestinal (GI) tract. In the present study, we investigated the distribution of Ano5 in the GI tracts of mice via reverse transcription-polymerase chain reaction (RT-PCR), Western blot and immunofluorescence analyses. The results indicated that Ano5 mRNA and protein are widely expressed in the esophagus, the stomach, the duodenum, the colon and the rectum but that Ano5 immunoreactivity was only detected in the mucosal layer, except for the muscular layer of the upper esophagus, which consists of skeletal muscle. In conclusion, our present results demonstrate for the first time the expression of Ano5 in the GI epithelium and in skeletal muscle in the esophagus. This novel finding facilitates clinical differential diagnosis and treatment. However, further investigation of the role of Ano5 in GI function is required.


Asunto(s)
Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Tracto Gastrointestinal/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Animales , Anoctaminas , Western Blotting , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Distrofia Muscular de Cinturas/genética , Mutación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcolema/metabolismo , Distribución Tisular
15.
Br J Pharmacol ; 168(1): 253-65, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22946960

RESUMEN

BACKGROUND AND PURPOSE: Ca(2+)-dependent Cl(-) secretion (CaCC) in airways and other tissues is due to activation of the Cl(-) channel TMEM16A (anoctamin 1). Earlier studies suggested that Ca(2+) -activated Cl(-) channels are regulated by membrane lipid inositol phosphates, and that 1-O-octyl-2-O-butyryl-myo-inositol 3,4,5,6-tetrakisphosphate octakis(propionoxymethyl) ester (INO-4995) augments CaCC. Here we examined whether TMEM16A is the target for INO-4995 and if the channel is regulated by inositol phosphates. EXPERIMENTAL APPROACH: The effects of INO-4995 on CaCC were examined in overexpressing HEK293, colonic and primary airway epithelial cells as well as Xenopus oocytes. We used patch clamping, double electrode voltage clamp and Ussing chamber techniques. KEY RESULTS: We found that INO-4995 directly activates a TMEM16A whole cell conductance of 6.1 ± 0.9 nS pF(-1) in overexpressing cells. The tetrakisphosphates Ins(3,4,5,6)P(4) or Ins(1,3,4,5)P(4) and enzymes controlling levels of InsP(4) or PIP(2) and PIP(3) had no effects on the magnitude or kinetics of TMEM16A currents. In contrast in Xenopus oocytes, human airways and colonic cells, which all express TMEM16A endogenously, Cl(-) currents were not acutely activated by INO-4995. However incubation with INO-4995 augmented 1.6- to 4-fold TMEM16A-dependent Cl(-) currents activated by ionomycin or ATP, while intracellular Ca(2+) signals were not affected. The potentiating effect of INO-4995 on transient ATP-activated TMEM16A-currents in cystic fibrosis (CF) airways was twice of that observed in non-CF airways. CONCLUSIONS AND IMPLICATIONS: These data indicate that TMEM16A is the target for INO-4995, although the mode of action appears different for overexpressed and endogenous channels. INO-4995 may be useful for the treatment of CF lung disease.


Asunto(s)
Canales de Cloruro/efectos de los fármacos , Canales de Cloruro/metabolismo , Fosfatos de Inositol/farmacología , Proteínas de Neoplasias/efectos de los fármacos , Animales , Anoctamina-1 , Bronquios/citología , Células Cultivadas , Fibrosis Quística , Regulador de Conductancia de Transmembrana de Fibrosis Quística/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células HEK293/efectos de los fármacos , Células HEK293/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Ionomicina/farmacología , Proteínas de Neoplasias/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Xenopus
16.
J Cell Sci ; 125(Pt 21): 4991-8, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22946059

RESUMEN

Anoctamin 1 (Ano1; TMEM16A) and anoctamin 2 (Ano2; TMEM16B) are novel Cl(-) channels transiently activated by an increase in intracellular Ca(2+). These channels are essential for epithelial Cl(-) secretion, smooth muscle peristalsis and olfactory signal transduction. They are central to inherited diseases and cancer and can act as heat sensors. Surprisingly, another member of this protein family, Ano6, operates as a Ca(2+)-activated phospholipid scramblase, and others were reported as intracellular proteins. It is therefore unclear whether anoctamins constitute a family of Ca(2+)-activated Cl(-) channels, or are proteins with heterogeneous functions. Using whole-cell patch clamping we demonstrate that Ano4-10 are all able to produce transient Ca(2+)-activated Cl(-) currents when expressed in HEK293 cells. Although some anoctamins (Ano1, 2, 4, 6, 7) were found to be well expressed in the plasma membrane, others (Ano8, 9, 10) show rather poor membrane expression and were mostly retained in the cytosol. The transient nature of the Cl(-) currents was demonstrated to be independent of intracellular Ca(2+) levels. We show that inactivation of Ano1 currents occurs in the continuous presence of elevated Ca(2+) concentrations, possibly by calmodulin-dependent kinase. The present results demonstrate that anoctamins are a family of Ca(2+)-activated Cl(-) channels, which also induce permeability for cations. They may operate as Cl(-) channels located in the plasma membrane or in intracellular compartments. These results increase our understanding of the physiological significance of anoctamins and their role in disease.


Asunto(s)
Canales de Cloruro/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Adenosina Trifosfato/fisiología , Anoctamina-1 , Anoctaminas , Calcio/metabolismo , Calcio/fisiología , Ionóforos de Calcio/farmacología , Señalización del Calcio , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Canales de Cloruro/fisiología , Citoplasma/metabolismo , Células HEK293 , Células HT29 , Humanos , Ionomicina/farmacología , Potenciales de la Membrana , Proteínas de la Membrana/fisiología , Anotación de Secuencia Molecular , Proteínas de Neoplasias/fisiología , Técnicas de Placa-Clamp , Proteínas de Transferencia de Fosfolípidos/fisiología , Transporte de Proteínas
17.
PLoS One ; 7(8): e41664, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22899999

RESUMEN

KCNMA1 encodes the α-subunit of the large conductance, voltage and Ca(2+)-activated (BK) potassium channel and has been reported as a target gene of genomic amplification at 10q22 in prostate cancer. To investigate the prevalence of the amplification in other human cancers, the copy number of KCNMA1 was analyzed by fluorescence-in-situ-hybridization (FISH) in 2,445 tumors across 118 different tumor types. Amplification of KCNMA1 was restricted to a small but distinct fraction of breast, ovarian and endometrial cancer with the highest prevalence in invasive ductal breast cancers and serous carcinoma of ovary and endometrium (3-7%). We performed an extensive analysis on breast cancer tissue microarrays (TMA) of 1,200 tumors linked to prognosis. KCNMA1 amplification was significantly associated with high tumor stage, high grade, high tumor cell proliferation, and poor prognosis. Immunofluorescence revealed moderate or strong KCNMA1 protein expression in 8 out of 9 human breast cancers and in the breast cancer cell line MFM223. KCNMA1-function in breast cancer cell lines was confirmed by whole-cell patch clamp recordings and proliferation assays, using siRNA-knockdown, BK channel activators such as 17ß-estradiol and the BK-channel blocker paxilline. Our findings revealed that enhanced expression of KCNMA1 correlates with and contributes to high proliferation rate and malignancy of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Células MCF-7 , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Interferencia de ARN , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
18.
Int J Biochem Cell Biol ; 44(11): 1897-900, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22710346

RESUMEN

Airways consist of a heterogeneous population of cells, comprising ciliated cells, Clara cells and goblet cells. Electrolyte secretion by the airways is necessary to produce the airway surface liquid that allows for mucociliary clearance of the lungs. Secretion is driven by opening of Cl(-) selective ion channels in the apical membrane of airway epithelial cells, through either receptor mediated increase in intracellular cAMP or cytosolic Ca(2+). Traditionally cAMP-dependent and Ca(2+)-dependent secretory pathways are regarded as independent. However, this concept has been challenged recently. With identification of the Ca(2+) activated Cl(-) channel TMEM16A (anoctamin 1) and with detailed knowledge of the cAMP-regulated cystic fibrosis transmembrane conductance regulator (CFTR), it has become possible to look more closely into this relationship.


Asunto(s)
Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteínas de la Membrana/metabolismo , Sistema Respiratorio/patología , Animales , Humanos , Modelos Biológicos
19.
Pflugers Arch ; 463(6): 819-27, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22526458

RESUMEN

Cystic fibrosis lung disease is caused by reduced Cl(-) secretion along with enhanced Na(+) absorption, leading to reduced airway surface liquid and compromised mucociliary clearance. Therapeutic strategies have been developed to activate cystic fibrosis transmembrane conductance regulator (CFTR) or to overcome enhanced Na(+) absorption by the epithelial Na(+) channel (ENaC). In a split-ubiquitin-based two-hybrid screening, we identified stress-associated ER protein 1 (SERP1)/ribosome-associated membrane protein 4 as a novel interacting partner for the ENaC ß-subunit. SERP1 is induced during cell stress and interacts with the molecular chaperone calnexin, thus controlling early biogenesis of membrane proteins. ENaC activity was measured in the human airway epithelial cell lines H441 and A549 and in voltage clamp experiments with ENaC-overexpressing Xenopus oocytes. We found that expression of SERP1 strongly inhibits amiloride-sensitive Na(+) transport. SERP1 coimmunoprecipitated and colocalized with ßENaC in the endoplasmic reticulum, together with the chaperone calnexin. In contrast to the inhibitory effects on ENaC, SERP1 appears to promote expression of CFTR. Taken together, SERP1 is a novel cochaperone and regulator of ENaC expression.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Proteínas de la Membrana/metabolismo , Oocitos/metabolismo , Mucosa Respiratoria/metabolismo , Estrés Fisiológico/fisiología , Animales , Calnexina/metabolismo , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Hipoxia/metabolismo , Oocitos/citología , Mucosa Respiratoria/citología , Xenopus laevis
20.
Exp Physiol ; 97(2): 184-92, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21908539

RESUMEN

Endogenous Ca(2+)-activated Cl(-) currents (CaCCs) are abundant and present in very different cell types. Very good evidence has been provided that endogenous CaCC is produced by anoctamin 1 (Ano1) and Ano2. Insight into the physiological role of anoctamins has been provided for Ano1, Ano2 and Ano6; however, the physiological role of the other seven members of the anoctamin family remains obscure. Anoctamins 1 and 2 may operate as individual Ca(2+)-sensitive channel proteins or may require accessory subunits for complete function. We find that overexpressed Ano1 has properties resembling all those of endogenous CaCCs, although with some noticeable biophysical and regulatory differences when compared with endogenous channels. Apart from Ano1 and Ano2, expression of Ano6 also produces a Cl(-) conductance. Depending on the cellular background, Ano6 currents may have variable properties. Anoctamins 1 and 6 are frequent in epithelial cells, often coexpressed together with Ano8, Ano9 and Ano10. Most available data on anoctamins were obtained from mouse tissues and from cultured cells, which may not be representative of native human tissues.


Asunto(s)
Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Animales , Calcio/metabolismo , Canales de Cloruro/biosíntesis , Epitelio/metabolismo , Humanos , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...