Littérature scientifique sur le sujet « Potassium channels, pancreatic cancer »

Créez une référence correcte selon les styles APA, MLA, Chicago, Harvard et plusieurs autres

Choisissez une source :

Consultez les listes thématiques d’articles de revues, de livres, de thèses, de rapports de conférences et d’autres sources académiques sur le sujet « Potassium channels, pancreatic cancer ».

À côté de chaque source dans la liste de références il y a un bouton « Ajouter à la bibliographie ». Cliquez sur ce bouton, et nous générerons automatiquement la référence bibliographique pour la source choisie selon votre style de citation préféré : APA, MLA, Harvard, Vancouver, Chicago, etc.

Vous pouvez aussi télécharger le texte intégral de la publication scolaire au format pdf et consulter son résumé en ligne lorsque ces informations sont inclues dans les métadonnées.

Articles de revues sur le sujet "Potassium channels, pancreatic cancer"

1

Remigante, Alessia, Paolo Zuccolini, Raffaella Barbieri, Loretta Ferrera, Rossana Morabito, Paola Gavazzo, Michael Pusch et Cristiana Picco. « NS-11021 Modulates Cancer-Associated Processes Independently of BK Channels in Melanoma and Pancreatic Duct Adenocarcinoma Cell Lines ». Cancers 13, no 23 (6 décembre 2021) : 6144. http://dx.doi.org/10.3390/cancers13236144.

Texte intégral
Résumé :
Potassium channels have emerged as regulators of carcinogenesis, thus introducing possible new therapeutic strategies in the fight against cancer. In particular, the large-conductance Ca2+-activated K+ channel, often referred to as BK channel, is involved in several cancer-associated processes. Here, we investigated the effects of different BK activators, NS-11021, NS-19504, and BMS-191011, in IGR39 (primary melanoma cell line) and Panc-1 (primary pancreatic duct carcinoma cell line), highly expressing the channel, and in IGR37 (metastatic melanoma cell line) that barely express BK. Our data showed that NS-11021 and NS-19504 potently activated BK channels in IGR39 and Panc-1 cells, while no effect on channel activation was detected in IGR37 cells. On the contrary, BK channel activator BMS-191011 was less effective. However, only NS-11021 showed significant effects in cancer-associated processes, such as cell survival, migration, and proliferation in these cancer cell lines. Moreover, NS-11021 led to an increase of intracellular Ca2+ concentration, independent of BK channel activation, thus complicating any interpretation of its role in the regulation of cancer-associated mechanisms. Overall, we conclude that the activation of the BK channel by itself is not sufficient to produce beneficial anti-cancer effects in the melanoma and PDAC cell lines examined. Importantly, our results raise an alarm flag regarding the use of presumably specific BK channel openers as anti-cancer agents.
Styles APA, Harvard, Vancouver, ISO, etc.
2

Iorio, Jessica, Lorenzo Antonuzzo, Emanuela Scarpi, Massimo D’Amico, Claudia Duranti, Luca Messerini, Clotilde Sparano et al. « Prognostic role of hERG1 Potassium Channels in Neuroendocrine Tumours of the Ileum and Pancreas ». International Journal of Molecular Sciences 23, no 18 (13 septembre 2022) : 10623. http://dx.doi.org/10.3390/ijms231810623.

Texte intégral
Résumé :
hERG1 potassium channels are widely expressed in human cancers of different origins, where they affect several key aspects of cellular behaviour. The present study was designed to evaluate the expression and clinical relevance of hERG1 protein in cancer tissues from patients suffering from neuroendocrine tumours (NETs) of ileal (iNETs) and pancreatic (pNETs) origin, with available clinicopathological history and follow-up. The study was carried out by immunohistochemistry with an anti-hERG1 monoclonal antibody. In a subset of samples, a different antibody directed against the hERG1/β1 integrin complex was also used. The analysis showed for the first time that hERG1 is expressed in human NETs originating from either the ileum or the pancreas. hERG1 turned out to have a prognostic value in NETs, showing (i) a statistically significant positive impact on OS of patients affected by ileal NETs, regardless the TNM stage; (ii) a statistically significant positive impact on OS of patients affected by aggressive (TNM stage IV) disease, either ileal or pancreatic; (iii) a trend to a negative impact on OS of patients affected by less aggressive (TNM stage I-III) disease, either ileal or pancreatic. Moreover, in order to evaluate whether ERG1 was functionally expressed in a cellular model of pNET, the INS1E rat insulinoma cell line was used, and it emerged that blocking ERG1 with a specific inhibitor of the channel (E4031) turned out in a significant reduction in cell proliferation.
Styles APA, Harvard, Vancouver, ISO, etc.
3

Manoli, Sagar, Stefano Coppola, Claudia Duranti, Matteo Lulli, Lara Magni, Nirmala Kuppalu, Nikolaj Nielsen et al. « The Activity of Kv 11.1 Potassium Channel Modulates F-Actin Organization During Cell Migration of Pancreatic Ductal Adenocarcinoma Cells ». Cancers 11, no 2 (23 janvier 2019) : 135. http://dx.doi.org/10.3390/cancers11020135.

Texte intégral
Résumé :
Cell migration exerts a pivotal role in tumor progression, underlying cell invasion and metastatic spread. The cell migratory program requires f-actin re-organization, generally coordinated with the assembly of focal adhesions. Ion channels are emerging actors in regulating cell migration, through different mechanisms. We studied the role of the voltage dependent potassium channel KV 11.1 on cell migration of pancreatic ductal adenocarcinoma (PDAC) cells, focusing on its effects on f-actin organization and dynamics. Cells were cultured either on fibronectin (FN) or on a desmoplastic matrix (DM) with the addition of a conditioned medium produced by pancreatic stellate cells (PSC) maintained in hypoxia (Hypo-PSC-CM), to better mimic the PDAC microenvironment. KV11.1 was essential to maintain stress fibers in a less organized arrangement in cells cultured on FN. When PDAC cells were cultured on DM plus Hypo-PSC-CM, KV11.1 activity determined the organization of cortical f-actin into sparse and long filopodia, and allowed f-actin polymerization at a high speed. In both conditions, blocking KV11.1 impaired PDAC cell migration, and, on cells cultured onto FN, the effect was accompanied by a decrease of basal intracellular Ca2+ concentration. We conclude that KV11.1 is implicated in sustaining pro-metastatic signals in pancreatic cancer, through a reorganization of f-actin in stress fibers and a modulation of filopodia formation and dynamics.
Styles APA, Harvard, Vancouver, ISO, etc.
4

Jiang, Shuheng, Lili Zhu, Jianyu Yang, Lipeng Hu, Jianren Gu, Xin Xing, Yongwei Sun et Zhigang Zhang. « Integrated expression profiling of potassium channels identifys KCNN4 as a prognostic biomarker of pancreatic cancer ». Biochemical and Biophysical Research Communications 494, no 1-2 (décembre 2017) : 113–19. http://dx.doi.org/10.1016/j.bbrc.2017.10.072.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
5

Wei, Mengyan, Pu Wang, Xiufang Zhu, Masaki Morishima, Yangong Liu, Mingqi Zheng, Gang Liu et al. « Electrophysiological evaluation of an anticancer drug gemcitabine on cardiotoxicity revealing down-regulation and modification of the activation gating properties in the human rapid delayed rectifier potassium channel ». PLOS ONE 18, no 2 (2 février 2023) : e0280656. http://dx.doi.org/10.1371/journal.pone.0280656.

Texte intégral
Résumé :
Gemcitabine is an antineoplastic drug commonly used in the treatment of several types of cancers including pancreatic cancer and non–small cell lung cancer. Although gemcitabine-induced cardiotoxicity is widely recognized, the exact mechanism of cardiac dysfunction causing arrhythmias remains unclear. The objective of this study was to electrophysiologically evaluate the proarrhythmic cardiotoxicity of gemcitabine focusing on the human rapid delayed rectifier potassium channel, hERG channel. In heterologous hERG expressing HEK293 cells (hERG-HEK cells), hERG channel current (IhERG) was reduced by gemcitabine when applied for 24 h but not immediately after the application. Gemcitabine modified the activation gating properties of the hERG channel toward the hyperpolarization direction, while inactivation, deactivation or reactivation gating properties were unaffected by gemcitabine. When gemcitabine was applied to hERG-HEK cells in combined with tunicamycin, an inhibitor of N-acetylglucosamine phosphotransferase, gemcitabine was unable to reduce IhERG or shift the activation properties toward the hyperpolarization direction. While a mannosidase I inhibitor kifunensine alone reduced IhERG and the reduction was even larger in combined with gemcitabine, kifunensine was without effect on IhERG when hERG-HEK cells were pretreated with gemcitabine for 24 h. In addition, gemcitabine down-regulated fluorescence intensity for hERG potassium channel protein in rat neonatal cardiomyocyte, although hERG mRNA was unchanged. Our results suggest the possible mechanism of arrhythmias caused by gemcitabine revealing a down-regulation of IhERG through the post-translational glycosylation disruption possibly at the early phase of hERG channel glycosylation in the endoplasmic reticulum that alters the electrical excitability of cells.
Styles APA, Harvard, Vancouver, ISO, etc.
6

Xu, Rui, Qiuyan Xu, Guanglei Huang, Xinhai Yin, Jianguo Zhu, Yikun Peng et Jukun Song. « Combined Analysis of the Aberrant Epigenetic Alteration of Pancreatic Ductal Adenocarcinoma ». BioMed Research International 2019 (28 décembre 2019) : 1–11. http://dx.doi.org/10.1155/2019/9379864.

Texte intégral
Résumé :
Background. Pancreatic ductal adenocarcinoma (PDAC) remains one of the most fatal malignancies due to its high morbidity and mortality. DNA methylation exerts a vital part in the development of PDAC. However, a mechanistic role of mutual interactions between DNA methylation and mRNA as epigenetic regulators on transcriptomic alterations and its correlation with clinical outcomes such as survival have remained largely uncovered in cancer. Therefore, elucidation of aberrant epigenetic alteration in the development of PDAC is an urgent problem to be solved. In this work, we conduct an integrative epigenetic analysis of PDAC to identify aberrant DNA methylation-driven cancer genes during the occurrence of cancer. Methods. DNA methylation matrix and mRNA profile were obtained from the TCGA database. The integration of methylation and gene expression datasets was analyzed using an R package MethylMix. The genes with hypomethylation/hypermethylation were further validated in the Kaplan–Meier analysis. The correlation analysis of gene expression and aberrant DNA methylation was also conducted. We performed a pathway analysis on aberrant DNG methylation genes identified by MethylMix criteria using ConsensusPathDB. Results. 188 patients with both methylation data and mRNA data were considered eligible. A mixture model was constructed, and differential methylation genes in normal and tumor groups using the Wilcoxon rank test was performed. With the inclusion criteria, 95 differential methylation genes were detected. Among these genes, 74 hypermethylation and 21 hypomethylation genes were found. The pathway analysis revealed an increase in hypermethylation of genes involved in ATP-sensitive potassium channels, Robo4, and VEGF signaling pathways crosstalk, and generic transcription pathway. Conclusion. Integrated analysis of the aberrant epigenetic alteration in pancreatic ductal adenocarcinoma indicated that differentially methylated genes could play a vital role in the occurrence of PDAC by bioinformatics analysis. The present work can help clinicians to elaborate on the function of differentially methylated expressed genes and pathways in PDAC. CDO1, GJD2, ID4, NOL4, PAX6, TRIM58, and ZNF382 might act as aberrantly DNA-methylated biomarkers for early screening and therapy of PDAC in the future.
Styles APA, Harvard, Vancouver, ISO, etc.
7

Li, Weiwei, Gregory C. Wilson, Magdalena Bachmann, Jiang Wang, Andrea Mattarei, Cristina Paradisi, Michael J. Edwards et al. « Inhibition of a Mitochondrial Potassium Channel in Combination with Gemcitabine and Abraxane Drastically Reduces Pancreatic Ductal Adenocarcinoma in an Immunocompetent Orthotopic Murine Model ». Cancers 14, no 11 (25 mai 2022) : 2618. http://dx.doi.org/10.3390/cancers14112618.

Texte intégral
Résumé :
Pancreas ductal adenocarcinoma (PDAC) is one the most aggressive cancers and associated with very high mortality, requiring the development of novel treatments. The mitochondrial voltage-gated potassium channel, Kv1.3 is emerging as an attractive oncologic target but its function in PDAC is unknown. Here, we evaluated the tissue expression of Kv1.3 in resected PDAC from 55 patients using immunohistochemistry (IHC) and show that all tumors expressed Kv1.3 with 60% of tumor specimens having high Kv1.3 expression. In Pan02 cells, the recently developed mitochondria-targeted Kv1.3 inhibitors PCARBTP and PAPTP strongly reduced cell survival in vitro. In an orthotopic pancreas tumor model (Pan02 cells injected into C57BL/6 mice) in immune-competent mice, injection of PAPTP or PCARBTP resulted in tumor reductions of 87% and 70%, respectively. When combined with clinically used Gemcitabine plus Abraxane (albumin-bound paclitaxel), reduction reached 95% and 80% without resultant organ toxicity. Drug-mediated tumor cell death occurred through the p38-MAPK pathway, loss of mitochondrial membrane potential, and oxidative stress. Resistant Pan02 clones to PCARBTP escaped cell death through upregulation of the antioxidant system. In contrast, tumor cells did not develop resistance to PAPTP. Our data show that Kv1.3 is highly expressed in resected human PDAC and the use of novel mitochondrial Kv1.3 inhibitors combined with cytotoxic chemotherapies might be a novel, effective treatment for PDAC.
Styles APA, Harvard, Vancouver, ISO, etc.
8

Huang, Xi, et Lily Yeh Jan. « Targeting potassium channels in cancer ». Journal of Cell Biology 206, no 2 (21 juillet 2014) : 151–62. http://dx.doi.org/10.1083/jcb.201404136.

Texte intégral
Résumé :
Potassium channels are pore-forming transmembrane proteins that regulate a multitude of biological processes by controlling potassium flow across cell membranes. Aberrant potassium channel functions contribute to diseases such as epilepsy, cardiac arrhythmia, and neuromuscular symptoms collectively known as channelopathies. Increasing evidence suggests that cancer constitutes another category of channelopathies associated with dysregulated channel expression. Indeed, potassium channel–modulating agents have demonstrated antitumor efficacy. Potassium channels regulate cancer cell behaviors such as proliferation and migration through both canonical ion permeation–dependent and noncanonical ion permeation–independent functions. Given their cell surface localization and well-known pharmacology, pharmacological strategies to target potassium channel could prove to be promising cancer therapeutics.
Styles APA, Harvard, Vancouver, ISO, etc.
9

Huang, Xi, et Lily Yeh Jan. « Targeting potassium channels in cancer ». Journal of General Physiology 144, no 2 (28 juillet 2014) : 1442OIA34. http://dx.doi.org/10.1085/jgp.1442oia34.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
10

Hayashi, Mikio, et Ivana Novak. « Molecular basis of potassium channels in pancreatic duct epithelial cells ». Channels 7, no 6 (2 novembre 2013) : 432–41. http://dx.doi.org/10.4161/chan.26100.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.

Thèses sur le sujet "Potassium channels, pancreatic cancer"

1

Li, Fangfang. « Regulation of pancreatic β-cell death and cancer cell migration by TPRM2 channels ». Thesis, University of Leeds, 2016. http://etheses.whiterose.ac.uk/13374/.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
2

Newton, Hannah S. « Potassium channels and adenosine signaling in T cells of head and neck cancer patients ». University of Cincinnati / OhioLINK, 2020. http://rave.ohiolink.edu/etdc/view?acc_num=ucin1603713656776019.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
3

Kondratska-Klymenko, Kateryna. « Role of calcium-permeable channels in pancreatic ductal adenocarcinoma resistance to chemotherapy ». Thesis, Lille 1, 2015. http://www.theses.fr/2015LIL10099/document.

Texte intégral
Résumé :
L'adénocarcinome pancréatique (PDAC) est la forme la plus fréquente de néoplasme de cet organe puisqu’il représente environ 90% de toutes les tumeurs pancréatiques et constitue l'une des principales causes de décès par cancer chez l’Homme. Le taux de survie à 5 ans n’est que de 6%. L'une des raisons à cela est que, au début du développement du cancer du pancréas, il n'y a pas de symptômes, et donc la majorité des cas sont diagnostiqués à des stades tardifs métastatiques ou invasifs pour lesquels une intervention chirurgicale n’est plus possible. Il a été montré que les cellules du cancer du pancréas présentent plusieurs mutations génétiques qui conduisent à la prolifération incontrôlée des cellules, ainsi qu’à l'évasion de l'apoptose. Les changements de concentration du Ca2+ cytosolique jouent un rôle central dans de nombreux processus cellulaires fondamentaux, et la perturbation des mécanismes de régulation de l'homéostasie du Ca2+ conduit à une grande variété de pathologies graves, dont le cancer. C’est notamment le cas pour les canaux calciques de type SOC, qui régulent une variété de processus cellulaires dépendants du calcium. Cependant, bien que le rôle du Ca2+ et des canaux calciques soit bien établi dans de nombreuses voies de signalisation de différents types cellulaires, les informations sur le rôle des canaux calciques dans le PDAC sont limitées. Donc, l'identification de la nature moléculaire ainsi que des fonctions des canaux calciques revêt une grande importance dans ces cellules car elle pourrait à termes fournir de nouvelles approches relatives au traitement du cancer du pancréas par le ciblage des processus dépendants du calcium
Pancreatic ductal adenocarcinoma (PDAC) representing the most prevalent pancreatic neoplasm accounting for about 90% of all pancreatic tumors, is one of the leading causes of cancer death in men and women. The current five-year relative survival rate is about 6% . One of the reasons of this is that early stage pancreatic cancer usually has no symptoms and thus the majority of cases are diagnosed at the late metastatic or invasive stages which are not suitable for surgery. Pancreatic cancer cells have been shown to exhibit a number of genetic mutations leading to uncontrolled cell proliferation, as well as evasion of programmed cell death (apoptosis). Changes in the cytosolic free Ca2+ concentration, play a central role in many fundamental cellular processes and disturbance of the Ca2+ homeostasis regulatory mechanisms leads to a vast variety of severe pathologies, including cancer. Among these, store-operated calcium channels (SOCs) have been shown to regulate a variety of calcium dependent cellular processes altered in different cancers. However, although the role of Ca2+ and calcium-permeable channels is well established in many signaling pathways in a variety of cell types, the information of the role of calcium-permeable channels in PDAC cells is limited. Therefore, identification of the molecular nature as well as functions of calcium-permeable channels in these cells is of great importance as it can reveal novel approaches for treating pancreatic cancer through targeting calcium-dependent processes
Styles APA, Harvard, Vancouver, ISO, etc.
4

Asher, Viren. « The expression of EAG and HERG potassium channels in ovarian cancer and their role in cell proliferation ». Thesis, University of Nottingham, 2013. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.594215.

Texte intégral
Résumé :
Background Ovarian cancer is the second most common cancer of the female genital tract in the United Kingdom (UK), accounting for 6% of female deaths due to cancer. This cancer is associated with poor survival and there is a need for new treatments in addition to existing chemotherapy to improve survival. Potassium (K) channels have shown to be promising therapeutic targets in the treatment of various cancers. We sought to determine the expression of EAG and HERG potassium channels in ovarian cancer and establish their role in cell proliferation. Material and Methods The Trent Research and Ethics Committee granted initial approval for the study. Informed consent was obtained from all patients undergoing surgery for ovarian cancer and oopherectomy for benign causes for their participation in the study. The tissues were prospectively collected and analysed anonymously. Immunoflurescence. Immunohistochemistry, Western blotting and Reverse transcriptase Polymerase chain reaction experiments were used to determine the expression of EAG and HERG potassium channels in ovarian cancer and normal ovaries. The effect of the EAG blockers (imipramine and clofilium) and HERG blockers (E-4031 and ergtoxin) on SK~OV-3 ovarian cancer cell line proliferation was assessed using the MTS assay with further investigation of their role in the cell cycle and apoptosis determined by flow cytometry. Results EAG and HERG potassium channels have significant (P<0.001) higher expression in patients with ovarian cancer compared to normal ovarian cells and high expression of EAG channels is significantly associated with poor survival (P=0.016) unlike HERG channel expression where there was no correlation with survival. There was also a significant association of EAG staining with high tumour grade (p=0.014) and presence of residual disease (p=0.011). Proliferation of SK-OV-3 cells was significantly (p<0.001) inhibited after treatment with voltage gated K+ channel blockers. There was significant inhibition of proliferation of SK-OV- 3 ovarian cancer cells by imipramine (p<0.001) and ergtoxin (p<0.05) at 72 hours of culture. Incubation of cells with ergtoxin led to the accumulation of cells in the S and G2IM phase while cells accumulated in S phase after incubation with E-403 1, with no effect on apoptosis.imipramine did not affect the cell cycle but increased the proportion ofSK-OV-3 cells undergoing early apoptosis. Conclusion Both EAG and HERG channels are expressed in ovarian cancer and have a role in cell proliferation. Higher expression of EAG channel is associated with poor prognosis suggesting its role as a poor prognostic marker in patients with ovarian cancer. HERG channels affect the cell cycle while EAG channels are implicated in the inhibition of apoptosis of ovarian cancer cells. EAG channels have the potential to be used as new therapeutic targets in patients with ovarian cancer with use of anti-EAG monoclonal antibody.
Styles APA, Harvard, Vancouver, ISO, etc.
5

Innamaa, Anni. « Expression and function of the two pore potassium (K2P) channels TREK-1, TREK-2 and TASK-3 in ovarian cancer ». Thesis, University of Nottingham, 2013. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.606812.

Texte intégral
Résumé :
Aberrant expression of potassium (K+) channels contributes to cancer cell proliferation and in certain circumstances channel blockade has been shown to inhibit cell proliferation. Two pore potassium (K2P) channels are the most recently identified group of K+ channels. K2P channels have been found to play a role in several cancers including prostate and breast cancer. We investigated the K2P channels TREK-1, TREK-2 and TASK-3, in ovarian cancer and normal ovaries and described the effect of channel blockade on cell proliferation, the cell cycle and apoptosis. Immunofluorescence confirmed expression in the cell lines (n=3) normal ovaries (n=4) and ovarian cancer (n=4). Western blotting quantified channel expression in normal ovaries (n=6) and cancer (n=22). There appeared to be a significant increase in expression of TREK-1 (P=O.0019) and TASK-3 (P=O.0047) in cancer when compared to normal ovaries. Immunohistochemistry further established expression in ovarian cancer (TREK-1 and -2, n=69) and normal ovaries (n=9) and in the TMA (TASK-3 n=230). Increased TASK-3 immunostaining conferred a significant survival advantage (P=O.001). There was a significant (P
Styles APA, Harvard, Vancouver, ISO, etc.
6

Vallejo, Gracia Albert. « Kv1.3 and Kv1.5 channels in leukocytes ». Doctoral thesis, Universitat de Barcelona, 2016. http://hdl.handle.net/10803/397797.

Texte intégral
Résumé :
Voltage dependent potassium channels are a group of plasma membrane ion channels with a key role in the immune system as the predominant ion channels controlling the resting membrane potential and tuning intracellular Ca2+ signaling in lymphocytes, monocytes, macrophages, and dendritic cells. Leukocytes present a limited Kv repertoire, including Kv1.3 and Kv1.5 channel isoforms. Kv1.3 is expressed in the immune system, and the blockade of this channel is associated with selective inhibition of T cell activation and proliferation. A functional Kv channel is an oligomeric complex composed of pore-forming and ancillary subunits. The KCNE gene family (KCNE1-5) is a novel group of modulatory Kv channel elements expressed in several tissues including leukocytes. KCNE peptides are small single spanning membrane proteins known to modulate Kv channels trafficking and biophysical properties. The hypothesis of the present PhD thesis entitled “Kv1.3 and Kv1.5 channels in leukocytes” was that changes in the channelosome composition by modulating the heterooligomeric combinations of the Kv1.3 channelosome control physiological and neoplastic cell growth as well as leukocyte responses. Evidence suggests that Kv channels are involved in cell differentiation and cell cycle control (because non-specific drugs, such as 4-AP and TEA, impaire proliferation), and they are also known to be remodeled during carcinogenesis. Thus, we elucidated the role of Kv1.3 and Kv1.5 channels in cell growth and their relationship with cancer, in models such as B lymphocytes and lymphomas (non-Hodgkin lymphomas), pancreatic ductal adenocarcinoma (PDAC) and glioblastomas. In spite of its significance, the mechanisms that regulate Kv1.3 and its role in the T cell activation are not well known. To that end, we analyzed the expression of KCNEs ancillary subunits upon different states of activation and proliferation of leukocytes (macrophages, T and B lymphocytes). In addition, recent data from our laboratory demonstrate that KCNE4, acting as a dominant negative ancillary subunit, physically interacts with Kv1.3 inhibiting K+ currents and retaining the channel intracellularly. Therefore, we studied the Kv1.3 modulation by the auxiliary subunit KCNE4 in leukocytes.
Styles APA, Harvard, Vancouver, ISO, etc.
7

Ahmed, Meftun. « Oscillatory Ca2+ signaling in glucose-stimulated murine pancreatic β-cells : Modulation by amino acids, glucagon, caffeine and ryanodine ». Doctoral thesis, Uppsala universitet, Institutionen för medicinsk cellbiologi, 2001. http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-1408.

Texte intégral
Résumé :
Oscillations in cytoplasmic Ca2+ concentration ([Ca2+]i) is the key signal in glucose-stimulated β-cells governing pulsatile insulin release. The glucose response of mouse β-cells is often manifested as slow oscillations and rapid transients of [Ca2+] i. In the present study, microfluorometric technique was used to evaluate the role of amino acids, glucagon, ryanodine and caffeine on the generation and maintenance of [Ca2+] i oscillations and transients in individual murine β-cells and isolated mouse pancreatic islets. The amino acids glycine, alanine and arginine, at around their physiological concentrations, transformed the glucose-induced slow oscillations of [Ca2+] i in isolated mouse β-cells into sustained elevation. Increased Ca2+ entry promoted the reappearance of the slow [Ca2+] i oscillations. The [Ca2+] i oscillations were more resistant to amino acid transformation in intact islets, supporting the idea that cellular interactions are important for maintaining the oscillatory activity. Individual rat β-cells responded to glucose stimulation with slow [Ca2+] i oscillations due to periodic entry of Ca2+ as well as with transients evoked by mobilization of intracellular stores. The [Ca2+] i oscillations in rat β-cells had a slightly lower frequency than those in mouse β-cells and were more easily transformed into sustained elevation in the presence of glucagon or caffeine. The transients of [Ca2+] i were more common in rat than in mouse β-cells and often appeared in synchrony also in cells lacking physical contact. Depolarization enhanced the generation of [Ca2+] i transients. In accordance with the idea that β-cells have functionally active ryanodine receptors, it was found that ryanodine sometimes restored oscillatory activity abolished by caffeine. However, the IP3 receptors are the major Ca2+ release channels both in β-cells from rats and mice. Single β-cells from ob/ob mice did not differ from those of lean controls with regard to frequency, amplitudes and half-widths of the slow [Ca2+] i oscillations. Nevertheless, there was an excessive firing of [Ca2+] i transients in the β-cells from the ob/ob mice, which was suppressed by leptin at close to physiological concentrations. The enhanced firing of [Ca2+] i transients in ob/ob mouse β-cells may be due to the absence of leptin and mediated by activation of the phospholipase C signaling pathway.
Styles APA, Harvard, Vancouver, ISO, etc.
8

Ahmed, Meftun. « Oscillatory Ca2+ signaling in glucose-stimulated murine pancreatic β-cells : Modulation by amino acids, glucagon, caffeine and ryanodine ». Doctoral thesis, Uppsala University, Department of Medical Cell Biology, 2001. http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-1408.

Texte intégral
Résumé :

Oscillations in cytoplasmic Ca2+ concentration ([Ca2+]i) is the key signal in glucose-stimulated β-cells governing pulsatile insulin release. The glucose response of mouse β-cells is often manifested as slow oscillations and rapid transients of [Ca2+] i. In the present study, microfluorometric technique was used to evaluate the role of amino acids, glucagon, ryanodine and caffeine on the generation and maintenance of [Ca2+] i oscillations and transients in individual murine β-cells and isolated mouse pancreatic islets. The amino acids glycine, alanine and arginine, at around their physiological concentrations, transformed the glucose-induced slow oscillations of [Ca2+] i in isolated mouse β-cells into sustained elevation. Increased Ca2+ entry promoted the reappearance of the slow [Ca2+] i oscillations. The [Ca2+] i oscillations were more resistant to amino acid transformation in intact islets, supporting the idea that cellular interactions are important for maintaining the oscillatory activity. Individual rat β-cells responded to glucose stimulation with slow [Ca2+] i oscillations due to periodic entry of Ca2+ as well as with transients evoked by mobilization of intracellular stores. The [Ca2+] i oscillations in rat β-cells had a slightly lower frequency than those in mouse β-cells and were more easily transformed into sustained elevation in the presence of glucagon or caffeine. The transients of [Ca2+] i were more common in rat than in mouse β-cells and often appeared in synchrony also in cells lacking physical contact. Depolarization enhanced the generation of [Ca2+] i transients. In accordance with the idea that β-cells have functionally active ryanodine receptors, it was found that ryanodine sometimes restored oscillatory activity abolished by caffeine. However, the IP3 receptors are the major Ca2+ release channels both in β-cells from rats and mice. Single β-cells from ob/ob mice did not differ from those of lean controls with regard to frequency, amplitudes and half-widths of the slow [Ca2+] i oscillations. Nevertheless, there was an excessive firing of [Ca2+] i transients in the β-cells from the ob/ob mice, which was suppressed by leptin at close to physiological concentrations. The enhanced firing of [Ca2+] i transients in ob/ob mouse β-cells may be due to the absence of leptin and mediated by activation of the phospholipase C signaling pathway.

Styles APA, Harvard, Vancouver, ISO, etc.
9

Serrano, Novillo Clara. « Biology of the cardiovascular Kv7.1 functional complex ». Doctoral thesis, Universitat de Barcelona, 2020. http://hdl.handle.net/10803/668686.

Texte intégral
Résumé :
Voltage gated K+ channels (Kv) are transmembrane proteins that allow the pass-thorugh of potassium ions, regulating the electrochemical gradient of the cell membrane. This way, they modulate several physiological processes, such as proliferation, migration or cell volume. Of particular interest in this dissertation is their role in excitable cells, were they control several key functions. The relevance of this ion channels is evidenced when mutations or alterations in the proper functioning of Kv channels causes severe pathologies, including cardiovascular or neuronal diseases, autoimmune affectations or cancer. Kv channels are tetramers of 4 α subunits with 6 transmembrane segments each one, that associate to form the pore and generate a functional channel. The wide functional diversity of currents is due to a vast number of modulations: heterotetramerization of α subunits, splicing variants, post-translational modifications or the association with regulatory subunits. The last ones include KCNE family, which co-assemble with the channel and modulate its electrophysiological, pharmacological or physiological properties. Kv7.1 associates with KCNE1 in cardiomyocytes to generate IKs cardiac repolarizing currents, in charge of finishing the cardiac action potential. Their assembly and traffic to the plasma membrane have been subject of discussion over the last years, with two opposite schools claiming an association early in the biogenesis versus a independent traffic to the plasma membrane, were both proteins would diffuse to assemble. We aimed in the present work to shed a light to this controversial topic. Kv channels have also been described in vascular smooth muscle, were they set the resting membrane potential and, therefore, control vascular tone. Kv7.1, Kv7.4 and Kv7.5 have been detected in different veins and arteries, were aberrations in their expression promote physiological alterations, but the specific role of each subunit remains unknown. In this scenario, the proposed objectives for the current PhD dissertation included the study of Kv7.1-KCNE1 complex, its assembly and traffic mechanisms. We hypothesized an unconventional secretion for the complex and suggest ER-PM junctions as the potential trafficking system. Therefore, we aim to characterize this structures and their implication in Kv7.1 membrane targeting. Finally, due to its implication in proliferation, their importance in cardiovascular system and their known role in some cancers, we studied the changes in the expression of Kv channels in endothelial-derived vascular tumors. We have been able to solve the traffic controversy of Kv7.1-KCNE1 complexes as they are not assembled early in their biogenesis. While KCNE1 is using the conventional secretion pathway, Kv7.1 takes an unconventional route that skips Golgi. Upon co-assembly, Kv7.1 redirects KCNE1 to this unconventional pathway. Moreover, we have proved that this non-conventional route are indeed ER-PM junctions, which also host the assembly of the complex. The molecular interactors of the channel during its ER-PM junction targeting have also been analysed during this PhD thesis, unravelling a complex and dynamic proteomic context. In addition, we have described for the first time the expression of Kv1.3, Kv1.5, Kv7.1 and Kv7.5 in endothelial cells of human veins and arteries. A remodelling of this composition is observed in different vascular cancers, related with the malignancy of the tumor in some of the cases.
Els canals de potassi dependents (Kv) regulen processos fisiològics molt importants, com la proliferació, la migració o el volum cel·lular. La seva rellevància es posa de manifest amb les diferents patologies associades a alteracions en la expressió dels canals, incloent malalties cardiovasculars, cerebrals, autoimmunes o càncer. Es tracta de proteïnes transmembrana formades per l’associació de 4 subunitats α que s’uneixen per formar el por. La gran varietat de diversitat funcional és deguda a la capacitat de heterotetramerització dels canals, variants d’splicing, modificacions post-traduccionals o la associació a subunitats reguladores KCNE, entre d’altres. En cardiomiòcits, Kv7.1 s’associa a KCNE1 per generar les corrents IKs, encarregades de la repolarització del potencial cardíac. La seva associació i tràfic són tema de debat des de fa anys, amb dues escoles defensant idees oposades. La primera, que les dues proteïnes s’associen en les fases inicials de la biogènesi; la segona, que trafiquen independent cap a la membrana, on difondran per trobar-se. Els Kv també s’han detectat a musculatura vascular llisa, on mantenen el potencial de repòs i controlen així el to vascular. Kv7.1, Kv7.4 i Kv7.5 es troben en diferents venes i arteries, on una expressió aberrant provoca alteracions fisiològiques. Tot i així, el seu paper concret encara es desconeix. En la present tesi doctoral hem comprovat que Kv7.1 i KCNE1 utilitzen vies diferents per arribar a la membrana plasmàtica. KCNE1 viatja per la via convencional, mentre que Kv7.1 utilitza una ruta no convencional que escapa del Golgi. Quan co-expressats, Kv7.1 redirigeix KCNE1 cap aquesta via alternativa. Hem demostrat que aquesta via són les ER-PM junctions, que també són el compartiment on la seva associació té lloc. Els interactors moleculars del canal durant el seu tràfic cap a ER-PM junctions també s’ha estudiat durant aquest treball. A més a més, hem descrit per primer cop l’expressió de Kv1.3, Kv1.5, Kv7.1 i Kv7.5 en l’endoteli de venes i artèries humanes. Hem vist un remodelatge en aquesta expressió en diferents càncers vasculars, en alguns casos relacionat amb la malignitat del tumor.
Styles APA, Harvard, Vancouver, ISO, etc.
10

Kaizik, Stephan Martin. « Analysis of mouse models of insulin secretion disorders ». Thesis, University of Oxford, 2010. http://ora.ox.ac.uk/objects/uuid:4d44b68a-a0a0-4c92-8809-00ddbfe3e636.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.

Chapitres de livres sur le sujet "Potassium channels, pancreatic cancer"

1

Camacho, Javier. « Ether à-go-go Potassium Channels ». Dans Encyclopedia of Cancer, 1–6. Berlin, Heidelberg : Springer Berlin Heidelberg, 2014. http://dx.doi.org/10.1007/978-3-642-27841-9_2028-8.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
2

Camacho, Javier. « Ether à-go-go Potassium Channels ». Dans Encyclopedia of Cancer, 1644–48. Berlin, Heidelberg : Springer Berlin Heidelberg, 2014. http://dx.doi.org/10.1007/978-3-662-46875-3_2028.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
3

Camacho, Javier. « Ether à-go-go Potassium Channels ». Dans Encyclopedia of Cancer, 1334–38. Berlin, Heidelberg : Springer Berlin Heidelberg, 2011. http://dx.doi.org/10.1007/978-3-642-16483-5_2028.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
4

Yee, Nelson S., et Rosemary K. Yee. « Ion Channels as Novel Pancreatic Cancer Biomarkers and Targets ». Dans New Advances on Disease Biomarkers and Molecular Targets in Biomedicine, 75–84. Totowa, NJ : Humana Press, 2013. http://dx.doi.org/10.1007/978-1-62703-456-2_5.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
5

Arcangeli, Annarosa. « Expression and Role of hERG Channels in Cancer Cells ». Dans The hERG Cardiac Potassium Channel : Structure, Function and Long QT Syndrome, 225–34. Chichester, UK : John Wiley & Sons, Ltd, 2008. http://dx.doi.org/10.1002/047002142x.ch17.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
6

Harding, E. A., C. Kane, R. F. L. James, N. J. M. London et M. J. Dunne. « Modulation of Three Types of Potassium Selective Channels by NAD and Other Pyridine Nucleotides in Human Pancreatic β-Cells ». Dans Advances in Experimental Medicine and Biology, 43–50. Boston, MA : Springer US, 1997. http://dx.doi.org/10.1007/978-1-4899-1819-2_6.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
7

Ganser, Katrin, Lukas Klumpp, Helmut Bischof, Robert Lukowski, Franziska Eckert et Stephan M. Huber. « Potassium Channels in Cancer ». Dans Handbook of Experimental Pharmacology. Berlin, Heidelberg : Springer Berlin Heidelberg, 2021. http://dx.doi.org/10.1007/164_2021_465.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
8

« Potassium Channels in Pancreatic P-Cells : Modulation, Pharmacology and their Role in the Regulation of Insulin Secretion ». Dans Potassium Channels And Their Modulators, 327–58. CRC Press, 1996. http://dx.doi.org/10.1201/9781482272840-20.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
9

Ningaraj, Nagendra S., et Divya Khaitan. « Targeting Potassium Channels for Drug Delivery to Brain Tumors ». Dans Frontiers in Anti-Cancer Drug Discovery, 231–46. BENTHAM SCIENCE PUBLISHERS, 2015. http://dx.doi.org/10.2174/9781681080581115050010.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
10

Yee, Nelson S. « TRPM8 Ion Channels as Potential Cancer Biomarker and Target in Pancreatic Cancer ». Dans Advances in Protein Chemistry and Structural Biology, 127–55. Elsevier, 2016. http://dx.doi.org/10.1016/bs.apcsb.2016.01.001.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.

Actes de conférences sur le sujet "Potassium channels, pancreatic cancer"

1

Coppola, Stefano, Claudia Duranti, Annarosa Arcangeli et Thomas Schmidt. « Abstract 183 : HERG1 potassium channels perturb the β1 integrins mediated force transduction machinery in pancreatic cancer ». Dans Proceedings : AACR Annual Meeting 2018 ; April 14-18, 2018 ; Chicago, IL. American Association for Cancer Research, 2018. http://dx.doi.org/10.1158/1538-7445.am2018-183.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
2

Coppola, S., C. Duranti, A. Arcangeli et T. Schmidt. « PO-268 The interaction of hERG1 potassium channels with integrin receptors perturbs the force transduction machinery in pancreatic cancer ». Dans Abstracts of the 25th Biennial Congress of the European Association for Cancer Research, Amsterdam, The Netherlands, 30 June – 3 July 2018. BMJ Publishing Group Ltd, 2018. http://dx.doi.org/10.1136/esmoopen-2018-eacr25.782.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
3

Breuer, Eun-Kyoung, Clodia Osipo, Jeremiah Zartman, Claire Wells, Michael Nishimura, Walter Jones et Saverio Gentile. « Abstract B38 : Exploring targeting potassium channels in cancer : A novel strategy for therapeutic intervention ». Dans Abstracts : AACR Precision Medicine Series : Opportunities and Challenges of Exploiting Synthetic Lethality in Cancer ; January 4-7, 2017 ; San Diego, CA. American Association for Cancer Research, 2017. http://dx.doi.org/10.1158/1538-8514.synthleth-b38.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
4

Ibrahim, S., A. Girault, L. Babin, M. Guéguinou, M. Potier-Cartereau, C. Vandier, G. Paintaud, T. Lecomte et W. Raoul. « PO-041 TNF pathway in metastatic colorectal cancer according to RAS status and implication of potassium channels ». Dans Abstracts of the 25th Biennial Congress of the European Association for Cancer Research, Amsterdam, The Netherlands, 30 June – 3 July 2018. BMJ Publishing Group Ltd, 2018. http://dx.doi.org/10.1136/esmoopen-2018-eacr25.86.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
5

Gupta, Bindiya, Puja Kumari, Shalini Rajaram, Rajarshi Kar, Priyanka Gogoi et Sandhya Jain. « 2022-RA-366-ESGO Calcium activated potassium channels (KCNMA1) as biomarker of pre invasive and invasive cervical cancer ». Dans ESGO 2022 Congress. BMJ Publishing Group Ltd, 2022. http://dx.doi.org/10.1136/ijgc-2022-esgo.866.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
6

Lo, Wing-Yee, Corinna Mohr, Friederike Steudel, Marjanka Schmidt, Douglas Easton, Reiner Hoppe, Werner Schroth, Peter Ruth, Robert Lukowski et Hiltrud Brauch. « Abstract 2030 : The role of genetic variation in calcium-activated potassium channels in breast cancer patients treated with tamoxifen ». Dans Proceedings : AACR 107th Annual Meeting 2016 ; April 16-20, 2016 ; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-2030.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
7

Das, Ronnie, Shubham Patle, Eric J. Seibel, Chris W. Burfeind et Saniel Lim. « Pathology in a tub step 2 : simple, rapid fabrication of curved, circular cross section millifluidic channels for biopsy preparation/3D imaging towards pancreatic cancer detection and diagnosis ». Dans Microfluidics, BioMEMS, and Medical Microsystems XVI, sous la direction de Bonnie L. Gray et Holger Becker. SPIE, 2018. http://dx.doi.org/10.1117/12.2291018.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
Nous offrons des réductions sur tous les plans premium pour les auteurs dont les œuvres sont incluses dans des sélections littéraires thématiques. Contactez-nous pour obtenir un code promo unique!