Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Pflugers Arch ; 476(6): 923-937, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38627262

RESUMO

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.


Assuntos
Canais Iônicos Sensíveis a Ácido , Meduloblastoma , Receptores Acoplados a Proteínas G , Humanos , Canais Iônicos Sensíveis a Ácido/metabolismo , Canais Iônicos Sensíveis a Ácido/genética , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Linhagem Celular Tumoral , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Concentração de Íons de Hidrogênio , Tamanho Celular , Morte Celular , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Cálcio/metabolismo , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia
2.
J Pineal Res ; 76(1): e12919, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37794846

RESUMO

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.


Assuntos
Canais de Cálcio , Melatonina , Humanos , Camundongos , Animais , Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Melatonina/farmacologia , Sódio/farmacologia , Potássio/farmacologia , Neurônios/metabolismo , Cálcio/metabolismo
3.
Pflugers Arch ; 475(9): 1073-1087, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37474775

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Neoplasias Cerebelares , Glioblastoma , Meduloblastoma , Humanos , Canais Iônicos Sensíveis a Ácido/metabolismo , Meduloblastoma/metabolismo , Glioblastoma/metabolismo , Neurônios/metabolismo , Linhagem Celular , Neoplasias Encefálicas/metabolismo , Cerebelo , Neoplasias Cerebelares/metabolismo , Microambiente Tumoral
4.
Pflugers Arch ; 475(3): 405-416, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36522586

RESUMO

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.


Assuntos
Glioblastoma , Humanos , Canais Iônicos Sensíveis a Ácido/genética , Canais Iônicos Sensíveis a Ácido/metabolismo , Concentração de Íons de Hidrogênio , Células-Tronco Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Microambiente Tumoral , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo
5.
BMC Biol ; 18(1): 143, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33059680

RESUMO

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.


Assuntos
Canal de Potássio Kv1.1/genética , Poli(ADP-Ribose) Polimerases/genética , Proteína Quinase C-delta/genética , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Animais , Células HEK293 , Células HeLa , Humanos , Canal de Potássio Kv1.1/metabolismo , Camundongos , Poli(ADP-Ribose) Polimerases/metabolismo , Proteína Quinase C-delta/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(17): E3507-E3515, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28396446

RESUMO

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.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Conotoxinas/química , Conotoxinas/toxicidade , Caramujo Conus/química , Gânglios Espinais/metabolismo , Músculo Esquelético/metabolismo , Mialgia/metabolismo , Neurônios/metabolismo , Animais , Gânglios Espinais/patologia , Camundongos , Músculo Esquelético/fisiologia , Mialgia/induzido quimicamente , Mialgia/patologia , Neurônios/patologia , Xenopus laevis
7.
J Cell Sci ; 125(Pt 21): 4991-8, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22946059

RESUMO

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.


Assuntos
Canais de Cloreto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Trifosfato de Adenosina/fisiologia , Anoctamina-1 , Anoctaminas , Cálcio/metabolismo , Cálcio/fisiologia , Ionóforos de Cálcio/farmacologia , Sinalização do Cálcio , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Canais de Cloreto/fisiologia , Citoplasma/metabolismo , Células HEK293 , Células HT29 , Humanos , Ionomicina/farmacologia , Potenciais da Membrana , Proteínas de Membrana/fisiologia , Anotação de Sequência Molecular , Proteínas de Neoplasias/fisiologia , Técnicas de Patch-Clamp , Proteínas de Transferência de Fosfolipídeos/fisiologia , Transporte Proteico
8.
Biochem Biophys Res Commun ; 451(2): 258-62, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25094048

RESUMO

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.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Trato Gastrointestinal/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Animais , Anoctaminas , Western Blotting , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcolema/metabolismo , Distribuição Tecidual
9.
ACS Chem Neurosci ; 14(14): 2487-2498, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37379568

RESUMO

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.


Assuntos
Canais Iônicos Sensíveis a Ácido , Chá , Ratos , Animais , Taninos Hidrolisáveis , Concentração de Íons de Hidrogênio
10.
Pflugers Arch ; 463(6): 819-27, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22526458

RESUMO

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.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Proteínas de Membrana/metabolismo , Oócitos/metabolismo , Mucosa Respiratória/metabolismo , Estresse Fisiológico/fisiologia , Animais , Calnexina/metabolismo , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Hipóxia/metabolismo , Oócitos/citologia , Mucosa Respiratória/citologia , Xenopus laevis
11.
Stem Cells ; 29(12): 2077-89, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22052697

RESUMO

Stem cell transplantation has been shown to improve functional outcome in degenerative and ischemic disorders. However, low in vivo survival and differentiation potential of the transplanted cells limits their overall effectiveness and thus clinical usage. Here we show that, after in vitro induction of neuronal differentiation and dedifferentiation, on withdrawal of extrinsic factors, mesenchymal stem cells (MSCs) derived from bone marrow, which have already committed to neuronal lineage, revert to a primitive cell population (dedifferentiated MSCs) retaining stem cell characteristics but exhibiting a reprogrammed phenotype distinct from their original counterparts. Of therapeutic interest, the dedifferentiated MSCs exhibited enhanced cell survival and higher efficacy in neuronal differentiation compared to unmanipulated MSCs both in vitro and in vivo, with significantly improved cognition function in a neonatal hypoxic-ischemic brain damage rat model. Increased expression of bcl-2 family proteins and microRNA-34a appears to be the important mechanism giving rise to this previously undefined stem cell population that may provide a novel treatment strategy with improved therapeutic efficacy.


Assuntos
Desdiferenciação Celular , Hipóxia-Isquemia Encefálica/terapia , Células-Tronco Mesenquimais/citologia , Neurônios/citologia , Animais , Diferenciação Celular , Sobrevivência Celular , Ventrículos Cerebrais/metabolismo , Técnicas de Cocultura , Peróxido de Hidrogênio/farmacologia , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Animais , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos
12.
FASEB J ; 25(3): 1058-68, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21115851

RESUMO

TMEM16A (anoctamin 1, Ano1), a member of a family of 10 homologous proteins, has been shown to form an essential component of Ca(2+)-activated Cl(-) channels. TMEM16A-null mice exhibit severe defects in epithelial transport along with tracheomalacia and death within 1 mo after birth. Despite its outstanding physiological significance, the mechanisms for activation of TMEM16A remain obscure. TMEM16A is activated on increase in intracellular Ca(2+), but it is unclear whether Ca(2+) binds directly to the channel or whether additional components are required. We demonstrate that TMEM16A is strictly membrane localized and requires cytoskeletal interactions to be fully activated. Despite the need for cytosolic ATP for full activation, phosphorylation by protein kinases is not required. In contrast, the Ca(2+) binding protein calmodulin appears indispensable and interacts physically with TMEM16A. Openers of small- and intermediate-conductance Ca(2+)-activated potassium channels known to interact with calmodulin, such as 1-EBIO, DCEBIO, or riluzole, also activated TMEM16A. These results reinforce the use of these compounds for activation of electrolyte secretion in diseases such as cystic fibrosis.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Cloretos/metabolismo , Fibrose Cística/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Anoctamina-1 , Benzimidazóis/farmacologia , Células COS , Cálcio/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Calmodulina/metabolismo , Chlorocebus aethiops , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Células HEK293 , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Fosforilação/fisiologia , Mucosa Respiratória/citologia , Traqueia/citologia
13.
Exp Physiol ; 97(2): 184-92, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21908539

RESUMO

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.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Animais , Cálcio/metabolismo , Canais de Cloreto/biossíntese , Epitélio/metabolismo , Humanos , Camundongos , Camundongos Knockout
14.
Cell Death Dis ; 13(8): 702, 2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-35961983

RESUMO

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.


Assuntos
Acidose , Glioblastoma , Canais Iônicos Sensíveis a Ácido/genética , Canais Iônicos Sensíveis a Ácido/metabolismo , Acidose/metabolismo , Animais , Glioblastoma/genética , Glioblastoma/metabolismo , Camundongos , Neurônios/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Células-Tronco/metabolismo , Microambiente Tumoral
15.
J Biol Chem ; 285(44): 33584-8, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20833712

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent monogenic cause of kidney failure, characterized by the development of renal cysts. ADPKD is caused by mutations of the polycystin-1 (PC1) or polycystin-2 (PC2) genes. PC2 encodes a Ca(2+)-permeable cation channel, and its dysfunction has been implicated in cyst development. The transcriptional coactivator with PDZ binding motif (TAZ) is required for the integrity of renal cilia. Its absence results in the development of renal cysts in a knock-out mouse model. TAZ directly interacts with PC2, and it has been suggested that another yet unidentified PDZ domain protein may be involved in the TAZ/PC2 interaction. Here we describe a novel interaction of TAZ with the multi-PDZ-containing PALS1-associated tight junction protein (PATJ). TAZ interacts with both the N-terminal PDZ domains 1-3 and the C-terminal PDZ domains 8-10 of PATJ, suggesting two distinct TAZ binding domains. We also show that the C terminus of PC2 strongly interacts with PDZ domains 8-10 and to a weaker extent with PDZ domains 1-3 of PATJ. Finally, we demonstrate that both TAZ and PATJ impair PC2 channel activity when co-expressed with PC2 in oocytes of Xenopus laevis. These results implicate TAZ and PATJ as novel regulatory elements of the PC2 channel and might thus be involved in ADPKD pathology.


Assuntos
Regulação da Expressão Gênica , Proteínas de Membrana/metabolismo , Núcleosídeo-Fosfato Quinase/metabolismo , Doenças Renais Policísticas/metabolismo , Canais de Cátion TRPP/genética , Junções Íntimas/metabolismo , Ativação Transcricional , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Oócitos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Canais de Cátion TRPP/metabolismo , Proteínas de Junções Íntimas , Xenopus laevis
16.
Pflugers Arch ; 461(5): 579-89, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21399895

RESUMO

Rotavirus infection is the most frequent cause for severe diarrhea in infants, killing more than 600,000 every year. The nonstructural protein NSP4 acts as a rotavirus enterotoxin, inducing secretory diarrhea without any structural organ damage. Electrolyte transport was assessed in the colonic epithelium from pups and adult mice using Ussing chamber recordings. Western blots and immunocytochemistry was performed in intestinal tissues from wild-type and TMEM16A knockout mice. Ion channel currents were recorded using patch clamp techniques. We show that the synthetic NSP4(114-135) peptide uses multiple pro-secretory pathways to induce diarrhea, by activating the recently identified Ca2+ -activated Cl- channel TMEM16A, and by inhibiting Na+ absorption by the epithelial Na+ channel ENaC and the Na+ /glucose cotransporter SGLT1. Activation of secretion and inhibition of Na+ absorption by NSP4(114-135), respectively, could be potently suppressed by wheat germ agglutinin which probably competes with NSP4(114-135) for binding to an unknown glycolipid receptor. The present paper gives a clue as to mechanisms of rotavirus-induced diarrhea and suggests wheat germ agglutinin as a simple and effective therapy.


Assuntos
Canais de Cloreto/fisiologia , Diarreia/etiologia , Glicoproteínas/farmacologia , Sódio/metabolismo , Toxinas Biológicas/farmacologia , Proteínas não Estruturais Virais/farmacologia , Envelhecimento , Animais , Anoctamina-1 , Colo/crescimento & desenvolvimento , Colo/metabolismo , Diarreia/virologia , Absorção Intestinal/efeitos dos fármacos , Camundongos , Infecções por Rotavirus
17.
Pflugers Arch ; 462(2): 195-208, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21607626

RESUMO

Endogenous Ca(2+)-activated Cl(-) channels (CaCC) demonstrate biophysical and pharmacological properties that are well represented in cells overexpressing anoctamin 1 (Ano 1, TMEM16A), a protein that has been identified recently as CaCC. Proteins of the anoctamin family (anoctamin 1-10, TMEM16A-K) are widely expressed. The number of reports demonstrating their physiological and clinical relevance is quickly rising. Anoctamins gain additional interest through their potential role in cell volume regulation and malignancy. Available data suggest that Ano 1 forms stable dimers and probably liaise with accessory proteins such as calmodulin or other anoctamins. In order to understand how anoctamins produce Ca(2+)-activated Cl(-) currents, it will be necessary to obtain better insight into their molecular structure, interactions with partner proteins, and mode of activation.


Assuntos
Canais de Cloreto/metabolismo , Animais , Cálcio/metabolismo , Canais de Cloreto/química , Canais de Cloreto/genética , Humanos , Proteínas de Membrana/metabolismo , Modelos Moleculares , Receptores de Superfície Celular/metabolismo
18.
Biol Chem ; 392(1-2): 125-34, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21194364

RESUMO

Two families of proteins, the bestrophins (Best) and the recently cloned TMEM16 proteins (anoctamin, Ano), recapitulate properties of Ca(2+)-activated Cl(-) currents. Best1 is strongly expressed in the retinal pigment epithelium and could have a function as a Ca(2+)-activated Cl(-) channel as well as a regulator of Ca(2+) signaling. It is also present at much lower levels in other cell types including epithelial cells, where it regulates plasma membrane localized Cl(-) channels by controlling intracellular Ca(2+) levels. Best1 interacts with important Ca(2+)-signaling proteins such as STIM1 and can interact directly with other Ca(2+)-activated Cl(-) channels such as TMEM16A. Best1 is detected in the endoplasmic reticulum (ER) where it shapes the dynamic ER structure and regulates cell proliferation, which could be important for renal cystogenesis. Ca(2+)-activated Cl(-) channels of the anoctamin family (TMEM16A) show biophysical and pharmacological properties that are typical for endogenous Ca(2+)-dependent Cl(-) channels. TMEM16 proteins are abundantly expressed and many reports demonstrate their physiological importance in epithelial as well as non-epithelial cells. These channels are also activated by cell swelling and can therefore control cell volume, proliferation and apoptosis. To fully understand the function and regulation of Ca(2+)-activated Cl(-) currents, it is necessary to appreciate that Best1 and TMEM16A are embedded in a protein network and that they probably operate in functional microdomains.


Assuntos
Canais de Cloreto/metabolismo , Células Epiteliais/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Anoctamina-1 , Bestrofinas , Células Epiteliais/química , Humanos
19.
Front Cell Neurosci ; 15: 668008, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33986647

RESUMO

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.

20.
J Biol Chem ; 284(42): 28571-8, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19654323

RESUMO

All vertebrate cells regulate their cell volume by activating chloride channels of unknown molecular identity, thereby activating regulatory volume decrease. We show that the Ca(2+)-activated Cl(-) channel TMEM16A together with other TMEM16 proteins are activated by cell swelling through an autocrine mechanism that involves ATP release and binding to purinergic P2Y(2) receptors. TMEM16A channels are activated by ATP through an increase in intracellular Ca(2+) and a Ca(2+)-independent mechanism engaging extracellular-regulated protein kinases (ERK1/2). The ability of epithelial cells to activate a Cl(-) conductance upon cell swelling, and to decrease their cell volume (regulatory volume decrease) was dependent on TMEM16 proteins. Activation of I(Cl,swell) was reduced in the colonic epithelium and in salivary acinar cells from mice lacking expression of TMEM16A. Thus TMEM16 proteins appear to be a crucial component of epithelial volume-regulated Cl(-) channels and may also have a function during proliferation and apoptotic cell death.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/fisiologia , Cloretos/química , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Animais , Anoctamina-1 , Apoptose , Cálcio/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Separação Celular , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA