45 results on '"Eag1"'
Search Results
2. Immunohistochemistry staining of Eag1 and p16/Ki-67 can help improve the management of patients with cervical intraepithelial Neoplasia after cold knife conversion.
- Author
-
Qiu, Shikang, Wang, Qiannan, Jiang, Huihui, and Feng, Limin
- Subjects
- *
CERVICAL intraepithelial neoplasia , *HUMAN papillomavirus , *IMMUNOHISTOCHEMISTRY , *LOGISTIC regression analysis , *RECEIVER operating characteristic curves , *P16 gene , *BIOMARKERS - Abstract
Background: Immunohistochemistry (IHC) is widely used in the management of patients with cervical intraepithelial neoplasia (CIN) but still has many limitations in clinical practice. We analyzed the correlation of new biomarkers with the severity of CIN and follow-up outcomes in patients after conization to improve the management of patients with CIN. Methods: IHC staining of Eag1 and p16/Ki-67 was performed on cervical tissue sections from 234 patients with suspected CIN2/3. After a series of follow-ups, including human papillomavirus (HPV) test and thinprep cytologic test (TCT) for 1–2 years, the outcomes were collected. IHC scores of biomarkers and follow-up results were used to analyze the correlation and assess the diagnostic efficiency of biomarkers. Results: The IHC staining intensity of Eag1 and p16/Ki-67 was significantly different from that of the CIN1-3 groups (p < 0.05). Eag1 expression scores were significantly different in the distribution between the two follow-up groups (p < 0.001). ROC curves based on the correlations between the follow-up outcomes and the Eag1 scores and IS of p16/ki-67 showed that Eag1 had a greater AUC (0.767 vs. 0.666). Logistic regression analysis of the combination of biomarkers revealed a greater AUC value than any single biomarker. Conclusions: Eag1 expression was significantly correlated with CIN grade and follow-up outcomes after conization. IHC staining of combinations of biomarkers of Eag1, p16 and Ki-67 may help us to improve the ability to identify risk groups with abnormal follow-up outcomes after treatment for CIN. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Overcoming challenges of HERG potassium channel liability through rational design: Eag1 inhibitors for cancer treatment.
- Author
-
Toplak, Žan, Hendrickx, Louise A., Abdelaziz, Reham, Shi, Xiaoyi, Peigneur, Steve, Tomašič, Tihomir, Tytgat, Jan, Peterlin‐Mašič, Lucija, and Pardo, Luis A.
- Subjects
POTASSIUM channels ,DRUG design ,ION channels ,POTASSIUM antagonists ,CANCER treatment ,VOLTAGE-gated ion channels ,TUMOR growth - Abstract
Two decades of research have proven the relevance of ion channel expression for tumor progression in virtually every indication, and it has become clear that inhibition of specific ion channels will eventually become part of the oncology therapeutic arsenal. However, ion channels play relevant roles in all aspects of physiology, and specificity for the tumor tissue remains a challenge to avoid undesired effects. Eag1 (KV10.1) is a voltage‐gated potassium channel whose expression is very restricted in healthy tissues outside of the brain, while it is overexpressed in 70% of human tumors. Inhibition of Eag1 reduces tumor growth, but the search for potent inhibitors for tumor therapy suffers from the structural similarities with the cardiac HERG channel, a major off‐target. Existing inhibitors show low specificity between the two channels, and screenings for Eag1 binders are prone to enrichment in compounds that also bind HERG. Rational drug design requires knowledge of the structure of the target and the understanding of structure–function relationships. Recent studies have shown subtle structural differences between Eag1 and HERG channels with profound functional impact. Thus, although both targets' structure is likely too similar to identify leads that exclusively bind to one of the channels, the structural information combined with the new knowledge of the functional relevance of particular residues or areas suggests the possibility of selective targeting of Eag1 in cancer therapies. Further development of selective Eag1 inhibitors can lead to first‐in‐class compounds for the treatment of different cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
4. Dronedarone blockage of the tumor-related Kv10.1 channel: a comparison with amiodarone.
- Author
-
Meléndez, T. A., Huanosta-Gutiérrez, A., Barriga-Montoya, C., González-Andrade, M., and Gómez-Lagunas, F.
- Subjects
- *
AMIODARONE , *DRUG interactions , *DRUG side effects , *POTASSIUM channels - Abstract
Kv10.1 (Eag1, or KCNH1) is a human potassium-selective channel associated with tumor development. In this work, we study the interaction of the drug dronedarone with Kv10.1. Dronedarone presents two chemical modifications aimed to lessen side effects produced by its parent molecule, the antiarrhythmic amiodarone. Hence, our observations are discussed within the framework of a previously reported interaction of amiodarone with Kv10.1. Additionally, we show new data regarding the interaction of amiodarone with the channels. We found that, unexpectedly, the effect of dronedarone on Kv10.1 differs both quantitatively and qualitatively to that of amiodarone. Among other observations, we found that dronedarone seems to be an open-pore blocker, in contrast to the reported behavior of amiodarone, which seems to inhibit from both open and closed states. Additionally, herein we provide evidence showing that, in spite of their chemical similarity, these molecules inhibit the K+ conductance by binding to non-overlapping, independent (non-allosterically related) sites. Also, we show that, while amiodarone inhibits the Cole-Moore shift, dronedarone is unable to inhibit this voltage-dependent characteristic of Kv10.1. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
5. A novel anti-cancer mechanism of Nutlin-3 through downregulation of Eag1 channel and PI3K/AKT pathway.
- Author
-
Wang, Xuzhao, Chen, Yafei, Liu, Hui, Guo, Shuai, Hu, Yufeng, Zhan, Yong, and An, Hailong
- Subjects
- *
HELA cells , *DOWNREGULATION , *CANCER cells , *CELL survival , *CELL lines - Abstract
Nutlin-3 shows a potent antitumor efficacy through downregulation of the cancerogenic ether à go-go 1 (Eag1) channel. However, the molecular mechanisms responsible for the regulation of Eag1 by Nutlin-3 in cancer cells remain unclear. In this study, we propose a novel anticancer mechanism of Nutlin-3, in which Nutlin-3 acts through the p53-Eag1-PI3K/AKT pathway. We first confirmed that Eag1 was downregulated through the activation of p53 by Nutlin-3. We then revealed that the inhibition of Eag1 electrophysiological function resulted in the decrease of viability, migration and invasion of HeLa cells. It is worth noting that the antitumor effect of Nutlin-3 was abolished in the Eag1 knockdown HeLa cell lines by siRNA. And Nutlin-3 can decrease the cell viability of H8 cells which were stably transfected with Eag1, but has no obvious inhibitory effect on blank H8 cells. Finally, we demonstrated that the decrease in Eag1 channel activity induced by Nutlin-3 treatment exerts anticancer activity by inhibiting the PI3K/AKT pathway. Our study therefore fills the gap between p53 pathway and its cellular function mediated by Eag1, shedding light on the new anti-cancer mechanism of Nutlin-3. • Eag1 current was recorded in HeLa cells for the first time. • The electrophysiological activity of Eag1 was reduced by Nutlin-3. • Anticancer molecular mechanism of Nutlin-3 depends on p53-Eag1-PI3K/AKT pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
6. Tetrandrine, a novel inhibitor of ether‐à‐go‐go‐1 (Eag1), targeted to cervical cancer development.
- Author
-
Wang, Xuzhao, Chen, Yafei, Li, Junwei, Guo, Shuai, Lin, Xiaoe, Zhang, Hailin, Zhan, Yong, and An, Hailong
- Subjects
- *
TETRANDRINE , *ETHER (Anesthetic) , *CERVICAL cancer , *CELL proliferation , *METASTASIS , *CANCER relapse - Abstract
Mortality‐to‐incidence ratios in patients with cancer are extremely high, positioning cancer as a major cause of death worldwide. Ether‐à‐go‐go‐1 (Eag1) is an ion channel that plays important roles in tumour proliferation, malignant transformation, invasion, metastasis, recurrence, and prognosis. Therefore, identifying potent and specific Eag1 channel inhibitors is crucial. In this study, we identified the first natural inhibitor of Eag1, the traditional Chinese medicine agent tetrandrine, and explored the underlying mechanism. Tetrandrine directly interacted with Eag1 and inhibited the currents in a concentration‐dependent manner (IC50 of 69.97 ± 5.2 μM), and the amino acids Ile 550, Thr 552, and Gln 557 in the Eag1 C‐linker domain were critical for tetrandrine's inhibitory effect. Moreover, tetrandrine reduced the proliferation of HeLa cells and Chinese hamster ovary (CHO) cells stably expressing Eag1 in a concentration‐dependent manner. Finally, tetrandrine (30 mg/kg/day) inhibited tumor growth in mice, demonstrating a 64.21% inhibitory rate of HeLa cell‐transplanted tumors. These results suggest that tetrandrine is a potent and selective Eag1 channel inhibitor, and could act as a leading compound in the development of therapies for Eag1 ion channel dysfunction‐induced diseases. We identified the first natural inhibitor of ether‐à‐go‐go‐1 (Eag1), the traditional Chinese medicine agent tetrandrine, and explored the underlying mechanism. Tetrandrine is a potent and selective Eag1 channel inhibitor, and could act as a leading compound in the development of therapies for Eag1 ion channel dysfunction‐induced diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
7. Eag1 channels as potential early-stage biomarkers of hepatocellular carcinoma
- Author
-
Chávez-López MG, Zúñiga-García V, Pérez-Carreón JI, Avalos-Fuentes A, Escobar Y, and Camacho J
- Subjects
Ion channels ,Eag1 ,Hepatocellular carcinoma ,Astemizole ,Diethylnitrosamine ,Medicine (General) ,R5-920 - Abstract
María de Guadalupe Chávez-López,1 Violeta Zúñiga-García,1 Julio Isael Pérez-Carreón,2 Arturo Avalos-Fuentes,3 Yesenia Escobar,4 Javier Camacho1 1Department of Pharmacology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 2Instituto Nacional de Medicina Genómica, 3Department of Physiology, Biophysics and Neuroscience, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 4Centro de Investigación Clínica Acelerada Sc, Mexico City, Mexico Abstract: Hepatocellular carcinoma (HCC) is a major cause of cancer death worldwide. HCC is usually asymptomatic at potential curative stages, and it has very poor prognosis if detected later. Thus, the identification of early biomarkers and novel therapies is essential to improve HCC patient survival. Ion channels have been proposed as potential tumor markers and therapeutic targets for several cancers including HCC. Especially, the ether à-go-go-1 (Eag1) voltage-gated potassium channel has been suggested as an early marker for HCC. Eag1 is overexpressed during HCC development from the cirrhotic and the preneoplastic lesions preceding HCC in a rat model. The channel is also overexpressed in human HCC. Astemizole has gained great interest as a potential anticancer drug because it targets several proteins involved in cancer including Eag1. Actually, in vivo studies have shown that astemizole may have clinical utility for HCC prevention and treatment. Here, we will review first some general aspects of HCC including the current biomarkers and therapies, and then we will focus on Eag1 channels as promising tools in the early diagnosis of HCC. Keywords: ion channels, Eag1, hepatocellular carcinoma, astemizole, diethylnitrosamine
- Published
- 2016
8. The involvement of Eag1 potassium channels and miR-34a in rotenone-induced death of dopaminergic SH-SY5Y cells.
- Author
-
HORST, CAMILA HILLESHEIM, TITZE-DE-ALMEIDA, RICARDO, and TITZE-DE-ALMEIDA, SIMONEIDE SOUZA
- Subjects
- *
ROTENONE , *BENZOPYRANS , *DIMETHYLTHIOUREA , *PHYSIOLOGICAL effects of potassium channels , *CELL survival - Abstract
The loss of dopaminergic neurons and the resultant motor impairment are hallmarks of Parkinson's disease. The SH-SY5Y cell line is a model of dopaminergic neurons, and allows for the study of dopaminergic neuronal injury. Previous studies have revealed changes in Ether à go-go 1 (Eag1) potassium channel expression during p53-induced SH-SY5Y apoptosis, and the regulatory involvement of microRNA-34a (miR-34a) was demonstrated. In the present study, the involvement of Eag1 and miR-34a in rotenone-induced SH-SY5Y cell injury was investigated. Rotenone is a neurotoxin, which is often used to generate models of Parkinson's disease, since it causes the death of nigrostriatal neurons by inducing intracellular aggregation of alpha synuclein and ubiquitin. In the present study, rotenone resulted in a dose-dependent decrease in cell viability, as revealed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and trypan blue cell counting assays. In addition, Eag1 was demonstrated to be constitutively expressed by SH-SY5Y cells, and involved in cell viability. Suppression of Eag1 with astemizole resulted in a dose-dependent decrease in cell viability, as revealed by MTT assay. Astemizole also enhanced the severity of rotenone-induced injury in SH-SY5Y cells. RNA interference against Eag1, using synthetic small interfering RNAs (siRNAs), corroborated this finding, as siRNAs potentiated rotenone-induced injury. Eag1-targeted siRNAs (kv10.1-3 or EAG1hum_287) resulted in a statistically significant 16.4-23.5% increase in vulnerability to rotenone. An increased number of apoptotic nuclei were observed in cells transfected with EAG1hum_287. Notably, this siRNA intensified rotenone-induced apoptosis, as revealed by an increase in caspase 3/7 activity. Conversely, a miR-34a inhibitor was demonstrated to exert neuroprotective effects. The viability of cells exposed to rotenone for 24 or 48 h and treated with miR-34a inhibitor was restored by 8.4-8.8%. In conclusion, Eag1 potassium channels and miR-34a are involved in the response to rotenone-induced injury in SH-SY5Y cells. The neuroprotective effect of mir-34a inhibitors merits further investigations in animal models of Parkinson's disease. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
9. Eag1 Voltage-Dependent Potassium Channels: Structure, Electrophysiological Characteristics, and Function in Cancer.
- Author
-
Wang, Xuzhao, Chen, Yafei, Zhang, Yuhong, Guo, Shuai, Mo, Li, An, Hailong, and Zhan, Yong
- Subjects
- *
POTASSIUM channels , *ELECTROPHYSIOLOGY , *MOLECULAR structure , *ADRENAL glands , *CANCER cell proliferation , *MYOBLASTS , *POTASSIUM metabolism , *PROTEIN metabolism , *ANIMALS , *CARRIER proteins , *POTASSIUM , *TUMORS - Abstract
Eag1 (ether-à-go-go-1), a member of the voltage-dependent potassium channel family, is expressed mainly in the brain, and at low levels in placenta, testis, and adrenal gland, and only transiently in myoblasts. Recently, several studies have suggested that Eag1 is selectively expressed in various tumor tissues. Eag1 plays important roles in tumor proliferation, malignant transformation, invasion, metastasis, recurrence, and prognosis. Therefore, it has become a new molecular target for tumor diagnosis, prognosis evaluation, and tumor-targeted therapy. This review provides information about the current progress in understanding Eag1 structure, electrophysiological characteristics, and role in cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
10. Eag1
- Author
-
Schwab, Manfred, editor
- Published
- 2017
- Full Text
- View/download PDF
11. Screening for Non-Pore-Binding Modulators of EAG K+ Channels.
- Author
-
Fernandes, Andreia S., Morais-Cabral, João H., and Harley, Carol A.
- Abstract
Members of the ether-à-go-go (EAG) family of voltage-gated K
+ channels are involved in several pathophysiological diseases, and there has been a great interest in screening for drugs that modulate the activity of these channels. Many drugs have been shown to bind in the pore of these channels, blocking ion flux and causing disease pathology. In this report, we present two independent screening campaigns in which we wanted to identify small molecules that bind to either the intracellular cytoplasmic amino terminal Per-Arnt-Sim (PAS) domain from the human EAG-related gene (ERG) channel or the amino or carboxy terminal globular domains from the mouse EAG1 channel, affecting their interaction. We report that in both cases, compounds were identified that showed weak, nonspecific binding. We suggest alternative routes should be pursued in future efforts to identify specific, high-affinity binders to these cytoplasmic domains. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
12. Molecular Dynamics-Derived Pharmacophore Model Explaining the Nonselective Aspect of KV10.1 Pore Blockers
- Author
-
Franci Merzel, Tihomir Tomašič, Žan Toplak, Lucija Peterlin Mašič, and Luis A. Pardo
- Subjects
QH301-705.5 ,hERG ,molekularna dinamika ,Antineoplastic Agents ,Molecular Dynamics Simulation ,udc:615.4:54:616-006 ,Ligands ,Article ,K$_V$10.1 inhibitors ,Catalysis ,Inorganic Chemistry ,Molecular dynamics ,Neoplasms ,Drug Discovery ,rak ,Potassium Channel Blockers ,Humans ,cancer ,Biology (General) ,Physical and Theoretical Chemistry ,Binding site ,QD1-999 ,Molecular Biology ,Eag1 ,Spectroscopy ,KV10.1 inhibitors ,Binding Sites ,biology ,pharmacophore ,Ligand ,Chemistry ,Organic Chemistry ,General Medicine ,Anticancer drug ,Ether-A-Go-Go Potassium Channels ,Potassium channel ,molecular dynamics ,Computer Science Applications ,HEK293 Cells ,zaviralci kalijevih kanalčkov ,farmakofor ,farmacevtska kemija ,Biophysics ,biology.protein ,Pharmacophore - Abstract
The KV10.1 voltage-gated potassium channel is highly expressed in 70% of tumors, and thus represents a promising target for anticancer drug discovery. However, only a few ligands are known to inhibit KV10.1, and almost all also inhibit the very similar cardiac hERG channel, which can lead to undesirable side-effects. In the absence of the structure of the KV10.1–inhibitor complex, there remains the need for new strategies to identify selective KV10.1 inhibitors and to understand the binding modes of the known KV10.1 inhibitors. To investigate these binding modes in the central cavity of KV10.1, a unique approach was used that allows derivation and analysis of ligand–protein interactions from molecular dynamics trajectories through pharmacophore modeling. The final molecular dynamics-derived structure-based pharmacophore model for the simulated KV10.1–ligand complexes describes the necessary pharmacophore features for KV10.1 inhibition and is highly similar to the previously reported ligand-based hERG pharmacophore model used to explain the nonselectivity of KV10.1 pore blockers. Moreover, analysis of the molecular dynamics trajectories revealed disruption of the π–π network of aromatic residues F359, Y464, and F468 of KV10.1, which has been reported to be important for binding of various ligands for both KV10.1 and hERG channels. These data indicate that targeting the KV10.1 channel pore is also likely to result in undesired hERG inhibition, and other potential binding sites should be explored to develop true KV10.1-selective inhibitors as new anticancer agents.
- Published
- 2021
- Full Text
- View/download PDF
13. Overcoming challenges of HERG potassium channel liability through rational design: Eag1 inhibitors for cancer treatment
- Author
-
Lucija Peterlin-Mašič, Reham Abdelaziz, Tihomir Tomašič, Xiaoyi Shi, Jan Tytgat, Steve Peigneur, Luis A. Pardo, Louise Antonia Hendrickx, and Žan Toplak
- Subjects
FUNCTIONAL EXPRESSION ,hERG ,Drug design ,HERG ,Chemistry, Medicinal ,ION-CHANNEL ,arrhythmia ,03 medical and health sciences ,0302 clinical medicine ,KAPPA-HEFUTOXIN 1 ,Neoplasms ,Drug Discovery ,medicine ,Humans ,cancer ,Pharmacology & Pharmacy ,Eag1 ,Ion channel ,030304 developmental biology ,Pharmacology ,RECTIFIER K+ CURRENT ,0303 health sciences ,Science & Technology ,ANTIARRHYTHMIC AGENT ,biology ,Chemistry ,A-GO-GO ,APOPTOSIS INDUCTION ,Rational design ,ABERRANT EXPRESSION ,Cancer ,medicine.disease ,potassium channels ,Ether-A-Go-Go Potassium Channels ,Potassium channel ,MOLECULAR DETERMINANTS ,3. Good health ,Cancer treatment ,Tumor progression ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Molecular Medicine ,REGULATES HUMAN ETHER ,rational drug design ,Life Sciences & Biomedicine - Abstract
Two decades of research have proven the relevance of ion channel expression for tumor progression in virtually every indication, and it has become clear that inhibition of specific ion channels will eventually become part of the oncology therapeutic arsenal. However, ion channels play relevant roles in all aspects of physiology, and specificity for the tumor tissue remains a challenge to avoid undesired effects. Eag1 (KV 10.1) is a voltage-gated potassium channel whose expression is very restricted in healthy tissues outside of the brain, while it is overexpressed in 70% of human tumors. Inhibition of Eag1 reduces tumor growth, but the search for potent inhibitors for tumor therapy suffers from the structural similarities with the cardiac HERG channel, a major off-target. Existing inhibitors show low specificity between the two channels, and screenings for Eag1 binders are prone to enrichment in compounds that also bind HERG. Rational drug design requires knowledge of the structure of the target and the understanding of structure-function relationships. Recent studies have shown subtle structural differences between Eag1 and HERG channels with profound functional impact. Thus, although both targets' structure is likely too similar to identify leads that exclusively bind to one of the channels, the structural information combined with the new knowledge of the functional relevance of particular residues or areas suggests the possibility of selective targeting of Eag1 in cancer therapies. Further development of selective Eag1 inhibitors can lead to first-in-class compounds for the treatment of different cancers. ispartof: MEDICINAL RESEARCH REVIEWS vol:42 issue:1 pages:183-226 ispartof: location:United States status: published
- Published
- 2021
14. Knockdown of Eag1 Expression by RNA Interference Increases Chemosensitivity to Cisplatin in Ovarian Cancer Cells.
- Author
-
Hui, Chen, Lan, Zhang, Yue-li, Lin, Li-lin, Hong, and Li-lin, Huang
- Subjects
- *
GENE expression , *RNA interference , *CISPLATIN , *CANCER cell analysis , *OVARIAN cancer , *HEALTH outcome assessment - Abstract
Ether á go-go 1 (Eag1) is frequently highly expressed in various malignant cancers and its excessive expression is correlated with poor prognosis in various cancers. However, the relationship of Eag1 expression with the clinical outcome of patients having ovarian cancer treated with cisplatin-based adjuvant chemotherapy is still unknown. In this study, we measured the expression of Eag1 in ovarian cancer and investigated the association between cisplatin chemosensitivity of ovarian cancer cells and Eag1 expression level. We demonstrate that decreased expression of Eag1 correlates with favorable prognosis in patients treated with cisplatin-based adjuvant chemotherapy and predicts higher cisplatin sensitivity in ovarian cancer cells. In vitro, knockdown of Eag1 by small interfering RNA facilitated the sensitivity of ovarian cancer cells (SKOV3 and TYK) to cisplatin-induced apoptosis via nuclear factor κ-light chain-enhancer of activated B cells (NF-κB) pathway. Furthermore, knockdown of Eag1 expression was associated with decreased expression of the P-glycoprotein without affecting multidrug resistance-associated protein 1 expression. Taken together, Eag1 may serve as a potential indicator to predict Eag1 chemosensitivity, and silencing Eag1 may represent a potential therapeutic strategy for ovarian cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
15. Astemizole-based anticancer therapy for hepatocellular carcinoma (HCC), and Eag1 channels as potential early-stage markers of HCC.
- Author
-
Guadalupe Chávez-López, María, Pérez-Carreón, Julio, Zuñiga-García, Violeta, Díaz-Chávez, José, Herrera, Luis, Caro-Sánchez, Claudia, Acuña-Macías, Isabel, Gariglio, Patricio, Hernández-Gallegos, Elizabeth, Chiliquinga, Andrea, and Camacho, Javier
- Abstract
Hepatocellular carcinoma (HCC) has very poor prognosis. Astemizole has gained great interest as a potential anticancer drug because it targets several proteins involved in cancer including the Eag1 ( ether à-go-go-1) potassium channel that is overexpressed in human HCC. Eag1 channels are regulated by cancer etiological factors and have been proposed as early tumor markers. Here, we found that HepG2 and HuH-7 HCC cells displayed Eag1 messenger RNA (mRNA) and protein expression, determined by real-time RT-PCR and immunochemistry, respectively. Astemizole inhibited human HCC cell proliferation (assessed by metabolic activity assay) and induced apoptosis (studied with flow cytometry) in both cell lines. The subcellular Eag1 protein localization was modified by astemizole in the HepG2 cells. The treatment with astemizole prevented diethylnitrosamine (DEN)-induced rat HCC development in vivo (followed by studying γ-glutamyl transpeptidase (GGT) activity). The Eag1 mRNA and protein levels were increased in most DEN-treated groups but decreased after astemizole treatment. GGT activity was decreased by astemizole. The Eag1 protein was detected in cirrhotic and dysplastic rat livers. Astemizole might have clinical utility for HCC prevention and treatment, and Eag1 channels may be potential early HCC biomarkers. These data provide significant basis to include astemizole in HCC clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
16. In vivo dual targeting of the oncogenic Ether-àgo-go-1 potassium channel by calcitriol and astemizole results in enhanced antineoplastic effects in breast tumors.
- Author
-
García-Quiroz, Janice, García-Becerra, Rocío, Santos-Martínez, Nancy, Barrera, David, Ordaz-Rosado, David, Avila, Euclides, Halhali, Ali, Villanueva, Octavio, Ibarra-Sánchez, Marı́a J., Esparza-López, José, Gamboa-Domínguez, Armando, Camacho, Javier, Larrea, Fernando, and Díaz, Lorenza
- Subjects
- *
BREAST tumor treatment , *TARGETED drug delivery , *CALCITRIOL , *ASTEMIZOLE , *POTASSIUM channels , *ANTINEOPLASTIC agents - Abstract
Background: The oncogenic ether-à-go-go-1 potassium channel (EAG1) activity and expression are necessary for cell cycle progression and tumorigenesis. The active vitamin D metabolite, calcitriol, and astemizole, a promising antineoplastic drug, target EAG1 by inhibiting its expression and blocking ion currents, respectively. We have previously shown a synergistic antiproliferative effect of calcitriol and astemizole in breast cancer cells in vitro, but the effect of this dual therapy in vivo has not been studied. Methods: In the present study, we explored the combined antineoplastic effect of both drugs in vivo using mice xenografted with the human breast cancer cell line T-47D and a primary breast cancer-derived cell culture (MBCDF). Tumor-bearing athymic female mice were treated with oral astemizole (50 mg/kg/day) and/or intraperitoneal injections of calcitriol (0.03 μg/g body weight twice a week) during 3 weeks. Tumor sizes were measured thrice weekly. For mechanistic insights, we studied EAG1 expression by qPCR and Western blot. The expression of Ki-67 and the relative tumor volume were used as indicators of therapeutic efficacy. Results: Compared to untreated controls, astemizole and calcitriol significantly reduced, while the coadministration of both drugs further suppressed, tumor growth (P < 0.05). In addition, the combined therapy significantly downregulated tumoral EAG1 and Ki-67 expression. Conclusions: The concomitant administration of calcitriol and astemizole inhibited tumor growth more efficiently than each drug alone, which may be explained by the blocking of EAG1. These results provide the bases for further studies aimed at testing EAG1-dual targeting in breast cancer tumors expressing both EAG1 and the vitamin D receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
17. KV10.1 K+-channel plasma membrane discrete domain partitioning and its functional correlation in neurons.
- Author
-
Jiménez-Garduño, Aura M., Mitkovski, Miso, Alexopoulos, Ioannis K., Sánchez, Araceli, Stühmer, Walter, Pardo, Luis A., and Ortega, Alicia
- Subjects
- *
POTASSIUM channels , *CELL membranes , *NEURONS , *CELL proliferation , *CANCER invasiveness , *CENTRAL nervous system physiology - Abstract
Abstract: KV10.1 potassium channels are implicated in a variety of cellular processes including cell proliferation and tumour progression. Their expression in over 70% of human tumours makes them an attractive diagnostic and therapeutic target. Although their physiological role in the central nervous system is not yet fully understood, advances in their precise cell localization will contribute to the understanding of their interactions and function. We have determined the plasma membrane (PM) distribution of the KV10.1 protein in an enriched mouse brain PM fraction and its association with cholesterol- and sphingolipid-rich domains. We show that the KV10.1 channel has two different populations in a 3:2 ratio, one associated to and another excluded from Detergent Resistant Membranes (DRMs). This distribution of KV10.1 in isolated PM is cholesterol- and cytoskeleton-dependent since alteration of those factors changes the relationship to 1:4. In transfected HEK-293 cells with a mutant unable to bind Ca2+/CaM to KV10.1 protein, Kv10.1 distribution in DRM/non-DRM is 1:4. Mean current density was doubled in the cholesterol-depleted cells, without any noticeable effects on other parameters. These results demonstrate that recruitment of the KV10.1 channel to the DRM fractions involves its functional regulation. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
18. Atomistic Insights of Calmodulin Gating of Complete Ion Channels
- Author
-
Bioquímica y biología molecular, Biokimika eta biologia molekularra, Núñez Viadero, Eider, Muguruza Montero, Arantza, Villarroel Muñoz, Álvaro, Bioquímica y biología molecular, Biokimika eta biologia molekularra, Núñez Viadero, Eider, Muguruza Montero, Arantza, and Villarroel Muñoz, Álvaro
- Abstract
Intracellular calcium is essential for many physiological processes, from neuronal signaling and exocytosis to muscle contraction and bone formation. Ca2+ signaling from the extracellular medium depends both on membrane potential, especially controlled by ion channels selective to K+, and direct permeation of this cation through specialized channels. Calmodulin (CaM), through direct binding to these proteins, participates in setting the membrane potential and the overall permeability to Ca2+. Over the past years many structures of complete channels in complex with CaM at near atomic resolution have been resolved. In combination with mutagenesis-function, structural information of individual domains and functional studies, different mechanisms employed by CaM to control channel gating are starting to be understood at atomic detail. Here, new insights regarding four types of tetrameric channels with six transmembrane (6TM) architecture, Eag1, SK2/SK4, TRPV5/TRPV6 and KCNQ1–5, and its regulation by CaM are described structurally. Different CaM regions, N-lobe, C-lobe and EF3/EF4-linker play prominent signaling roles in different complexes, emerging the realization of crucial non-canonical interactions between CaM and its target that are only evidenced in the full-channel structure. Different mechanisms to control gating are used, including direct and indirect mechanical actuation over the pore, allosteric control, indirect effect through lipid binding, as well as direct plugging of the pore. Although each CaM lobe engages through apparently similar alpha-helices, they do so using different docking strategies. We discuss how this allows selective action of drugs with great therapeutic potential.
- Published
- 2020
19. Atomistic Insights of Calmodulin Gating of Complete Ion Channels
- Author
-
Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), Eusko Jaurlaritza, Núñez, Eider, Muguruza-Montero, Arantza, Villarroel, Álvaro, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), Eusko Jaurlaritza, Núñez, Eider, Muguruza-Montero, Arantza, and Villarroel, Álvaro
- Abstract
Intracellular calcium is essential for many physiological processes, from neuronal signaling and exocytosis to muscle contraction and bone formation. Ca2+ signaling from the extracellular medium depends both on membrane potential, especially controlled by ion channels selective to K+, and direct permeation of this cation through specialized channels. Calmodulin (CaM), through direct binding to these proteins, participates in setting the membrane potential and the overall permeability to Ca2+. Over the past years many structures of complete channels in complex with CaM at near atomic resolution have been resolved. In combination with mutagenesis-function, structural information of individual domains and functional studies, different mechanisms employed by CaM to control channel gating are starting to be understood at atomic detail. Here, new insights regarding four types of tetrameric channels with six transmembrane (6TM) architecture, Eag1, SK2/SK4, TRPV5/TRPV6 and KCNQ1–5, and its regulation by CaM are described structurally. Different CaM regions, N-lobe, C-lobe and EF3/EF4-linker play prominent signaling roles in different complexes, emerging the realization of crucial non-canonical interactions between CaM and its target that are only evidenced in the full-channel structure. Different mechanisms to control gating are used, including direct and indirect mechanical actuation over the pore, allosteric control, indirect effect through lipid binding, as well as direct plugging of the pore. Although each CaM lobe engages through apparently similar alpha-helices, they do so using different docking strategies. We discuss how this allows selective action of drugs with great therapeutic potential.
- Published
- 2020
20. Hippocampal ether-à-go-go1 potassium channels blockade: Effects in the startle reflex and prepulse inhibition.
- Author
-
Issy, A.C., Fonseca, J.R., Pardo, L.A., Stühmer, W., and Del Bel, E.A.
- Subjects
- *
HIPPOCAMPUS physiology , *POTASSIUM antagonists , *STARTLE reaction , *DENTATE gyrus , *PHYSIOLOGICAL effects of potassium channels - Abstract
Highlights: [•] Eag1 K+ channels blockade in the dentate gyrus of the hippocampus did not modify apomorphine-disruptive effects in the PPI. [•] Dentate gyrus surgery inducing decreased startle response was restored by the Eag1 antibody. [•] The role of Eag1 K+ channels in the startle response merits further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
21. Ether- à- go- go 1 (Eag1) Potassium Channel Expression in Dopaminergic Neurons of Basal Ganglia is Modulated by 6-Hydroxydopamine Lesion.
- Author
-
Ferreira, N., Mitkovski, M., Stühmer, W., Pardo, L., and Del Bel, E.
- Subjects
- *
POTASSIUM channels , *ETHER (Anesthetic) , *DOPAMINERGIC neurons , *BASAL ganglia , *6-Hydroxydopamine , *DOPAMINE , *NEUROTOXIC agents , *LABORATORY rats - Abstract
The ether à go- go (Eag) gene encodes the voltage-gated potassium (K) ion channel Kv10.1, whose function still remains unknown. As dopamine may directly affect K channels, we evaluated whether a nigrostriatal dopaminergic lesion induced by the neurotoxin 6-hydroxydopamine (6-OHDA) would alter Eag1-K channel expression in the rat basal ganglia and related brain regions. Male Wistar rats received a microinjection of either saline or 6-OHDA (unilaterally) into the medial forebrain bundle. The extent of the dopaminergic lesion induced by 6-OHDA was evaluated by apomorphine-induced rotational behavior and by tyrosine hydroxylase (TH) immunoreactivity. The 6-OHDA microinjection caused a partial or complete lesion of dopaminergic cells, as well as a reduction of Eag1+ cells in a manner proportional to the extent of the lesion. In addition, we observed a decrease in TH immunoreactivity in the ipsilateral striatum. In conclusion, the expression of the Eag1-K-channel throughout the nigrostriatal pathway in the rat brain, its co-localization with dopaminergic cells and its reduction mirroring the extent of the lesion highlight a physiological circuitry where the functional role of this channel can be investigated. The Eag1-K channel expression in dopaminergic cells suggests that these channels are part of the diversified group of ion channels that generate and maintain the electrophysiological activity pattern of dopaminergic midbrain neurons. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
22. Eag 1, Eag 2 and Kcnn3 gene brain expression of isolated reared rats Martin et al.
- Author
-
Martin, S., Lino-de-Oliveira, C., Joca, S. R. L., Weffort de Oliveira, R., Echeverry, M. B., Da Silva, C. A., Pardo, L., Stühmer, W., and Bel, E. D.
- Subjects
- *
GENE expression , *SEROTONIN , *MESSENGER RNA , *POTASSIUM channels , *TYROSINE - Abstract
The Eag1 and Eag2, voltage-dependent potassium channels, and the small-conductance calcium-activated potassium channel (Kcnn3) are highly expressed in limbic regions of the brain, where their function is still unknown. Eag1 co-localizes with tyrosine hydroxilase enzyme in the substantia nigra and ventral tegmental area. Kcnn3 deficiency leads to enhanced serotonergic and dopaminergic neurotransmission accompanied by distinct alterations in emotional behaviors. As exposure to stress is able to change the expression and function of several ion channels, suggesting that they might be involved in the consequences of stress, we aimed at investigating Eag 1, Eag2 and Kcnn3 mRNA expression in the brains of rats submitted to isolation rearing. As the long-lasting alterations in emotional and behavioral regulation after stress have been related to changes in serotonergic neurotransmission, expressions of serotonin Htr1a and Htr2a receptors in male Wistar rats' brain were also investigated. Rats were reared in isolation or in groups of five for nine weeks after weaning. Isolated and socially reared rats were tested for exploratory activity in the open field test for 5 min and brains were processed for reverse-transcription coupled to quantitative polymerase chain reaction (qRT-PCR). Isolated reared rats showed decreased exploratory activity in the open field. Compared to socially reared rats, isolated rats showed reduced Htr2a mRNA expression in the striatum and brainstem and reduced Eag2 mRNA expression in all examined regions except cerebellum. To our knowledge, this is the first work to show that isolation rearing can change Eag2 gene expression in the brain. The involvement of this channel in stress-related behaviors is discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
23. Eag1对骨肉瘤细胞增殖、迁移与侵袭的 影响及其机制.
- Author
-
袁忠, 胡俊, 雷家维, 付璐珩, 俞曾强, and 汪海燕
- Abstract
目的探讨Eag1对骨肉瘤细胞增殖、迁移与侵袭的影响及其机制。方法将骨肉瘤Saos-2细胞随机分 为Eag1沉默组和对照组,分别转染Eag1 siRNA及scrambk。分别采用荧光定量PCR法和Weston blotting法检测 两组Eag1 mRNA和蛋白相对表达量;MTT法检测两组培养24、48、72、96及120 h的细胞增殖能力(以OD值表 示),细胞划痕试验检测两组细胞迁移能力(以划痕愈合率表示),Transwell侵袭试验检测两组细胞侵袭能力(以进 人膜下的细胞数量表示);采用Western blotting法检测两组STAT3蛋白相对表达量结果Eag1沉默组与对照组 Eag1 mRNA相对表达量分别为0.50±0.04、1.00±0.12,Eag1蛋ft相对表达量分别为0.30 ± 0.06、1.00 ±0.10,两 组比较P均< 0.05。Eag1沉默组接种24、48、72、96及120 h的0D值均低于对照组(P <0.05或<0.01)。Eag1沉 默组和对照组划痕愈合率分别为42. 34% ±4. 22%、95. 12% ± 2. 32%,进人膜下的细胞数量分别为(352 ± 32)、 (642 ±34)个,STAT3蛋白相对表达量分别为0. 79 ± 0. 15、1 . 64 ± 0. 23,两组比较P <0. 05或<0. 01。结论Eag1 沉默可抑制骨肉瘤细胞的增殖、迁移及侵袭;下调STAT3蛋白表达可能是其作用机制. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
24. Inducing Polyclonal Eag1-Specific Antibodies by Vaccination with a Linear Epitope Immunogen and Its Relation to Breast Tumorigenesis
- Author
-
Li, Zhandong, Zhu, Ketong, Gong, Xin, Vasilescu, Steven, Sun, Yu, Hong, Kaiqing, Li, Hao, Li, Lin, and Shan, Yaming
- Published
- 2017
- Full Text
- View/download PDF
25. The involvement of Eag1 potassium channels and miR-34a in rotenone-induced death of dopaminergic SH-SY5Y cells
- Author
-
Simoneide Souza Titze-de-Almeida, Ricardo Titze-de-Almeida, and Camila Hillesheim Horst
- Subjects
0301 basic medicine ,Cancer Research ,SH-SY5Y ,Cell Survival ,Caspase 3 ,Biology ,Transfection ,Biochemistry ,Neuroprotection ,rotenone ,03 medical and health sciences ,chemistry.chemical_compound ,RNA interference ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,Humans ,neurodegenerative diseases ,MTT assay ,Gene Silencing ,RNA, Small Interfering ,Eag1 ,Molecular Biology ,Cell Death ,Dopaminergic Neurons ,Dopaminergic ,Articles ,Rotenone ,Astemizole ,Immunohistochemistry ,Ether-A-Go-Go Potassium Channels ,Cell biology ,MicroRNAs ,030104 developmental biology ,Oncology ,chemistry ,Apoptosis ,Molecular Medicine ,030217 neurology & neurosurgery ,Intracellular - Abstract
The loss of dopaminergic neurons and the resultant motor impairment are hallmarks of Parkinson's disease. The SH-SY5Y cell line is a model of dopaminergic neurons, and allows for the study of dopaminergic neuronal injury. Previous studies have revealed changes in Ether à go-go 1 (Eag1) potassium channel expression during p53-induced SH-SY5Y apoptosis, and the regulatory involvement of microRNA-34a (miR-34a) was demonstrated. In the present study, the involvement of Eag1 and miR-34a in rotenone-induced SH-SY5Y cell injury was investigated. Rotenone is a neurotoxin, which is often used to generate models of Parkinson's disease, since it causes the death of nigrostriatal neurons by inducing intracellular aggregation of alpha synuclein and ubiquitin. In the present study, rotenone resulted in a dose-dependent decrease in cell viability, as revealed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and trypan blue cell counting assays. In addition, Eag1 was demonstrated to be constitutively expressed by SH-SY5Y cells, and involved in cell viability. Suppression of Eag1 with astemizole resulted in a dose-dependent decrease in cell viability, as revealed by MTT assay. Astemizole also enhanced the severity of rotenone-induced injury in SH-SY5Y cells. RNA interference against Eag1, using synthetic small interfering RNAs (siRNAs), corroborated this finding, as siRNAs potentiated rotenone-induced injury. Eag1-targeted siRNAs (kv10.1-3 or EAG1hum_287) resulted in a statistically significant 16.4–23.5% increase in vulnerability to rotenone. An increased number of apoptotic nuclei were observed in cells transfected with EAG1hum_287. Notably, this siRNA intensified rotenone-induced apoptosis, as revealed by an increase in caspase 3/7 activity. Conversely, a miR-34a inhibitor was demonstrated to exert neuroprotective effects. The viability of cells exposed to rotenone for 24 or 48 h and treated with miR-34a inhibitor was restored by 8.4–8.8%. In conclusion, Eag1 potassium channels and miR-34a are involved in the response to rotenone-induced injury in SH-SY5Y cells. The neuroprotective effect of mir-34a inhibitors merits further investigations in animal models of Parkinson's disease.
- Published
- 2017
- Full Text
- View/download PDF
26. Atomistic Insights of Calmodulin Gating of Complete Ion Channels
- Author
-
Eider Nuñez, Alvaro Villarroel, Arantza Muguruza-Montero, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), and Eusko Jaurlaritza
- Subjects
calmodulin ,Potassium Channels ,Calmodulin ,Allosteric regulation ,Kv10 ,Kv7 ,Review ,Gating ,Ion Channels ,Catalysis ,Exocytosis ,Calcium in biology ,Inorganic Chemistry ,lcsh:Chemistry ,KCNQ ,Transient Receptor Potential Channels ,Allosteric Regulation ,Protein Domains ,SK2 ,SK4 ,Humans ,M-current ,Calcium Signaling ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Eag1 ,Spectroscopy ,Ion channel ,Membrane potential ,biology ,Chemistry ,Organic Chemistry ,General Medicine ,Transmembrane protein ,Computer Science Applications ,lcsh:Biology (General) ,lcsh:QD1-999 ,biology.protein ,Biophysics ,TRPV5 ,TRPV6 - Abstract
© 2020 by the authors, Intracellular calcium is essential for many physiological processes, from neuronal signaling and exocytosis to muscle contraction and bone formation. Ca2+ signaling from the extracellular medium depends both on membrane potential, especially controlled by ion channels selective to K+, and direct permeation of this cation through specialized channels. Calmodulin (CaM), through direct binding to these proteins, participates in setting the membrane potential and the overall permeability to Ca2+. Over the past years many structures of complete channels in complex with CaM at near atomic resolution have been resolved. In combination with mutagenesis-function, structural information of individual domains and functional studies, different mechanisms employed by CaM to control channel gating are starting to be understood at atomic detail. Here, new insights regarding four types of tetrameric channels with six transmembrane (6TM) architecture, Eag1, SK2/SK4, TRPV5/TRPV6 and KCNQ1–5, and its regulation by CaM are described structurally. Different CaM regions, N-lobe, C-lobe and EF3/EF4-linker play prominent signaling roles in different complexes, emerging the realization of crucial non-canonical interactions between CaM and its target that are only evidenced in the full-channel structure. Different mechanisms to control gating are used, including direct and indirect mechanical actuation over the pore, allosteric control, indirect effect through lipid binding, as well as direct plugging of the pore. Although each CaM lobe engages through apparently similar alpha-helices, they do so using different docking strategies. We discuss how this allows selective action of drugs with great therapeutic potential., The Government of the Autonomous Community of the Basque Country (IT1165–19) and the Spanish Ministry of Economy, Industry and Competitiveness (RTI2018–097839-B-100) provided financial support for this work. E.N. and A.M-M. were supported by predoctoral contracts of the Basque Government.
- Published
- 2020
27. Molecular Dynamics-Derived Pharmacophore Model Explaining the Nonselective Aspect of K V 10.1 Pore Blockers.
- Author
-
Toplak, Žan, Merzel, Franci, Pardo, Luis A., Peterlin Mašič, Lucija, and Tomašič, Tihomir
- Subjects
- *
POTASSIUM channels , *MOLECULAR dynamics , *PROTEIN-ligand interactions , *BINDING sites , *MOLECULAR interactions , *ANTINEOPLASTIC agents - Abstract
The KV10.1 voltage-gated potassium channel is highly expressed in 70% of tumors, and thus represents a promising target for anticancer drug discovery. However, only a few ligands are known to inhibit KV10.1, and almost all also inhibit the very similar cardiac hERG channel, which can lead to undesirable side-effects. In the absence of the structure of the KV10.1–inhibitor complex, there remains the need for new strategies to identify selective KV10.1 inhibitors and to understand the binding modes of the known KV10.1 inhibitors. To investigate these binding modes in the central cavity of KV10.1, a unique approach was used that allows derivation and analysis of ligand–protein interactions from molecular dynamics trajectories through pharmacophore modeling. The final molecular dynamics-derived structure-based pharmacophore model for the simulated KV10.1–ligand complexes describes the necessary pharmacophore features for KV10.1 inhibition and is highly similar to the previously reported ligand-based hERG pharmacophore model used to explain the nonselectivity of KV10.1 pore blockers. Moreover, analysis of the molecular dynamics trajectories revealed disruption of the π–π network of aromatic residues F359, Y464, and F468 of KV10.1, which has been reported to be important for binding of various ligands for both KV10.1 and hERG channels. These data indicate that targeting the KV10.1 channel pore is also likely to result in undesired hERG inhibition, and other potential binding sites should be explored to develop true KV10.1-selective inhibitors as new anticancer agents. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
28. Tumor cell-selective apoptosis induction through targeting of KV10.1 via bifunctional TRAIL antibody
- Author
-
Pardo Luis A, Stühmer Walter, and Hartung Franziska
- Subjects
KV10.1 ,Eag1 ,scFv62-TRAIL ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The search for strategies to target ion channels for therapeutic applications has become of increasing interest. Especially, the potassium channel KV10.1 (Ether-á-go-go) is attractive as target since this surface protein is virtually not detected in normal tissue outside the central nervous system, but is expressed in approximately 70% of tumors from different origins. Methods We designed a single-chain antibody against an extracellular region of KV10.1 (scFv62) and fused it to the human soluble TRAIL. The KV10.1-specific scFv62 antibody -TRAIL fusion protein was expressed in CHO-K1 cells, purified by chromatography and tested for biological activity. Results Prostate cancer cells, either positive or negative for KV10.1 were treated with the purified construct. After sensitization with cytotoxic drugs, scFv62-TRAIL induced apoptosis only in KV10.1-positive cancer cells, but not in non-tumor cells, nor in tumor cells lacking KV10.1 expression. In co-cultures with KV10.1-positive cancer cells the fusion protein also induced apoptosis in bystander KV10.1-negative cancer cells, while normal prostate epithelial cells were not affected when present as bystander. Conclusions KV10.1 represents a novel therapeutic target for cancer. We could design a strategy that selectively kills tumor cells based on a KV10.1-specific antibody.
- Published
- 2011
- Full Text
- View/download PDF
29. Astemizole-based anticancer therapy for hepatocellular carcinoma (HCC), and Eag1 channels as potential early-stage markers of HCC
- Author
-
de Guadalupe Chávez-López, María, Pérez-Carreón, Julio Isael, Zuñiga-García, Violeta, Díaz-Chávez, José, Herrera, Luis A., Caro-Sánchez, Claudia Haydee, Acuña-Macías, Isabel, Gariglio, Patricio, Hernández-Gallegos, Elizabeth, Chiliquinga, Andrea Jazmín, and Camacho, Javier
- Published
- 2015
- Full Text
- View/download PDF
30. Atomistic Insights of Calmodulin Gating of Complete Ion Channels.
- Author
-
Núñez, Eider, Muguruza-Montero, Arantza, and Villarroel, Alvaro
- Subjects
- *
CALMODULIN , *ION channels , *INTRACELLULAR calcium , *MEMBRANE potential , *CARRIER proteins , *ALLOSTERIC regulation - Abstract
Intracellular calcium is essential for many physiological processes, from neuronal signaling and exocytosis to muscle contraction and bone formation. Ca2+ signaling from the extracellular medium depends both on membrane potential, especially controlled by ion channels selective to K+, and direct permeation of this cation through specialized channels. Calmodulin (CaM), through direct binding to these proteins, participates in setting the membrane potential and the overall permeability to Ca2+. Over the past years many structures of complete channels in complex with CaM at near atomic resolution have been resolved. In combination with mutagenesis-function, structural information of individual domains and functional studies, different mechanisms employed by CaM to control channel gating are starting to be understood at atomic detail. Here, new insights regarding four types of tetrameric channels with six transmembrane (6TM) architecture, Eag1, SK2/SK4, TRPV5/TRPV6 and KCNQ1–5, and its regulation by CaM are described structurally. Different CaM regions, N-lobe, C-lobe and EF3/EF4-linker play prominent signaling roles in different complexes, emerging the realization of crucial non-canonical interactions between CaM and its target that are only evidenced in the full-channel structure. Different mechanisms to control gating are used, including direct and indirect mechanical actuation over the pore, allosteric control, indirect effect through lipid binding, as well as direct plugging of the pore. Although each CaM lobe engages through apparently similar alpha-helices, they do so using different docking strategies. We discuss how this allows selective action of drugs with great therapeutic potential. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
31. Eag1 channels as potential early-stage biomarkers of hepatocellular carcinoma
- Author
-
Julio Israel Pérez-Carreón, Javier Camacho, Arturo Avalos-Fuentes, María de Guadalupe Chávez-López, Yesenia Escobar, and Violeta Zúñiga-García
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Poor prognosis ,diethylnitrosamine ,Usually asymptomatic ,Review ,Gastroenterology ,03 medical and health sciences ,Rheumatology ,Internal medicine ,medicine ,Immunology and Allergy ,Pharmacology (medical) ,astemizole ,Stage (cooking) ,neoplasms ,Eag1 ,030102 biochemistry & molecular biology ,business.industry ,Cancer ,ion channels ,Patient survival ,hepatocellular carcinoma ,medicine.disease ,Anticancer drug ,digestive system diseases ,Oncology ,Astemizole ,Hepatocellular carcinoma ,Cancer research ,business ,medicine.drug - Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer death worldwide. HCC is usually asymptomatic at potential curative stages, and it has very poor prognosis if detected later. Thus, the identification of early biomarkers and novel therapies is essential to improve HCC patient survival. Ion channels have been proposed as potential tumor markers and therapeutic targets for several cancers including HCC. Especially, the ether a-go-go-1 (Eag1) voltage-gated potassium channel has been suggested as an early marker for HCC. Eag1 is overexpressed during HCC development from the cirrhotic and the preneoplastic lesions preceding HCC in a rat model. The channel is also overexpressed in human HCC. Astemizole has gained great interest as a potential anticancer drug because it targets several proteins involved in cancer including Eag1. Actually, in vivo studies have shown that astemizole may have clinical utility for HCC prevention and treatment. Here, we will review first some general aspects of HCC including the current biomarkers and therapies, and then we will focus on Eag1 channels as promising tools in the early diagnosis of HCC.
- Published
- 2016
32. In vivo dual targeting of the oncogenic Ether-à-go-go-1 potassium channel by calcitriol and astemizole results in enhanced antineoplastic effects in breast tumors
- Author
-
Rocío García-Becerra, Maŕa J Ibarra-Sánchez, Octavio Villanueva, Nancy Santos-Martínez, Armando Gamboa-Domínguez, David Barrera, David Ordaz-Rosado, Janice García-Quiroz, Euclides Avila, Javier Camacho, Fernando Larrea, José Esparza-López, Ali Halhali, and Lorenza Díaz
- Subjects
Cancer Research ,Calcitriol ,medicine.medical_treatment ,Mice, Nude ,Antineoplastic Agents ,Breast Neoplasms ,Pharmacology ,medicine.disease_cause ,Calcitriol receptor ,Targeted therapy ,Mice ,Breast cancer ,In vivo ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Genetics ,Animals ,Humans ,Vitamin D ,EAG1 ,biology ,business.industry ,Drug Synergism ,Astemizole ,medicine.disease ,Ether-A-Go-Go Potassium Channels ,Oncology ,Vitamin D receptor ,Ki-67 ,biology.protein ,Female ,business ,Carcinogenesis ,Neoplasm Transplantation ,medicine.drug ,Research Article - Abstract
Background The oncogenic ether-à-go-go-1 potassium channel (EAG1) activity and expression are necessary for cell cycle progression and tumorigenesis. The active vitamin D metabolite, calcitriol, and astemizole, a promising antineoplastic drug, target EAG1 by inhibiting its expression and blocking ion currents, respectively. We have previously shown a synergistic antiproliferative effect of calcitriol and astemizole in breast cancer cells in vitro, but the effect of this dual therapy in vivo has not been studied. Methods In the present study, we explored the combined antineoplastic effect of both drugs in vivo using mice xenografted with the human breast cancer cell line T-47D and a primary breast cancer-derived cell culture (MBCDF). Tumor-bearing athymic female mice were treated with oral astemizole (50 mg/kg/day) and/or intraperitoneal injections of calcitriol (0.03 μg/g body weight twice a week) during 3 weeks. Tumor sizes were measured thrice weekly. For mechanistic insights, we studied EAG1 expression by qPCR and Western blot. The expression of Ki-67 and the relative tumor volume were used as indicators of therapeutic efficacy. Results Compared to untreated controls, astemizole and calcitriol significantly reduced, while the coadministration of both drugs further suppressed, tumor growth (P
- Published
- 2014
33. HANR promotes lymphangiogenesis of hepatocellular carcinoma via secreting miR-296 exosome and regulating EAG1/VEGFA signaling in HDLEC cells.
- Author
-
Shi Y, Yang X, Xue X, Sun D, Cai P, Song Q, Zhang B, and Qin L
- Subjects
- Base Sequence, Carcinoma, Hepatocellular pathology, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Liver Neoplasms genetics, Liver Neoplasms pathology, MicroRNAs genetics, Models, Biological, Neoplasm Invasiveness, RNA, Long Noncoding genetics, Signal Transduction, Carcinoma, Hepatocellular genetics, Endothelial Cells metabolism, Ether-A-Go-Go Potassium Channels metabolism, Exosomes metabolism, Lymphangiogenesis genetics, MicroRNAs metabolism, RNA, Long Noncoding metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
The long noncoding RNA HANR has been shown to be involved in the progression of hepatocellular carcinoma (HCC). However, the underlying mechanism of HCC-associated long noncoding RNA (HANR)-regulated HCC metastasis and lymphangiogenesis has not been elucidated. RT-qPCR and Western blot methods were utilized to detect the gene expressions. Interaction of HANR with miR-296 was predicted by a bioinformatic program and validated by a dual-luciferase reporter assay. For the functional experiment, a transwell invasion assay was utilized to examine the invasion abilities of HepG2 and Huh-7 cells. The lymphatic vessel formation assay was used to show the HCC-associated lymphatic vessel formation ability of human dermal lymphatic endothelial cells (HDLEC). HANR was shown to directly bind to miR-296, and miR-296 downregulated HANR expression in HepG2 cells. Then, we observed that miR-296 inhibitor transfection in shHANR HCC cells could promote lymphatic vessel formation and invasion of HDLEC cells compared with shHANR HCC cells. EAG1 or VEGFA overexpression in HDLEC cells rescued lymphatic vessel formation and invasion in HDLEC cells coincubated with the medium of HepG2 cells expressing shHANR or miR-296 mimic. Ultimately, HANR knockdown and miR-296 mimic led to a significant decrease in the EAG1 and VEGFA expression levels in HepG2 cells. Here, we reveal a novel molecular mechanism in which the HANR/miR-296/EAG1/VEGF axis is responsible for the lymphangiogenesis of HCC cells. Our findings provide more insights into developing therapeutical or diagnostic methods by targeting HANR., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
34. Kν10.1 K⁺ - channel plasma membrane discrete domain partitioning and its functional correlation in neurons
- Author
-
Ioannis Alexopoulos, Alicia Ortega, Luis A. Pardo, Miso Mitkovski, Walter Stühmer, Aura Matilde Jiménez-Garduño, and Araceli Sánchez
- Subjects
Proteolipid protein 1 ,hERG ,Blotting, Western ,Detergents ,Biophysics ,Real-Time Polymerase Chain Reaction ,Biochemistry ,OLIG2 ,Mice ,Membrane Microdomains ,Ca2+/calmodulin-dependent protein kinase ,Cyclodextrin ,Animals ,Humans ,RNA, Messenger ,Eag1 ,Cytoskeleton ,Neurons ,biology ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Membrane ,Membrane ,Transfection ,Cell Biology ,Potassium channel ,Ether-A-Go-Go Potassium Channels ,Myelin basic protein ,Cell biology ,Electrophysiology ,Mice, Inbred C57BL ,Cholesterol ,HEK293 Cells ,KV10.1 ,Potassium Channels, Voltage-Gated ,biology.protein ,Female - Abstract
KV10.1 potassium channels are implicated in a variety of cellular processes including cell proliferation and tumour progression. Their expression in over 70% of human tumours makes them an attractive diagnostic and therapeutic target. Although their physiological role in the central nervous system is not yet fully understood, advances in their precise cell localization will contribute to the understanding of their interactions and function. We have determined the plasma membrane (PM) distribution of the KV10.1 protein in an enriched mouse brain PM fraction and its association with cholesterol- and sphingolipid-rich domains. We show that the KV10.1 channel has two different populations in a 3:2 ratio, one associated to and another excluded from Detergent Resistant Membranes (DRMs). This distribution of KV10.1 in isolated PM is cholesterol- and cytoskeleton-dependent since alteration of those factors changes the relationship to 1:4. In transfected HEK-293 cells with a mutant unable to bind Ca2+/CaM to KV10.1 protein, Kv10.1 distribution in DRM/non-DRM is 1:4. Mean current density was doubled in the cholesterol-depleted cells, without any noticeable effects on other parameters. These results demonstrate that recruitment of the KV10.1 channel to the DRM fractions involves its functional regulation.
- Published
- 2014
35. Increased expression of the oncotarget Eag1 in brain metastasis compared to their corresponding primary carcinomas
- Author
-
Martinez, R, Rohde, V, Reichmann, A, Martin, S, and Pardo, L
- Subjects
ddc: 610 ,oncotarget ,brain metastasis ,610 Medical sciences ,Medicine ,Eag1 - Abstract
Objective: Eag1, a voltage gated potassium channel is expressed in the brain, participating in the control of cell excitability. In addition, it has been implicated in malignant transformation and progression in a variety of human malignancies and in cancer cell lines. In this analysis, we aimed to [for full text, please go to the a.m. URL], 65. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC)
- Published
- 2014
- Full Text
- View/download PDF
36. Hippocampal ether-à-go-go1 potassium channels blockade: effects in the startle reflex and prepulse inhibition
- Author
-
A.C. Issy, J. R. Fonseca, Walter Stühmer, Luis A. Pardo, and E.A. Del Bel
- Subjects
Male ,Apomorphine ,Dopamine ,Neuroscience(all) ,Hippocampus ,Hippocampal formation ,HIPOCAMPU DE ANIMAL (EFEITOS DE DROGAS) ,Moro reflex ,Animals ,Rats, Wistar ,Eag1 ,Kv10.1 ,Prepulse inhibition ,Chemistry ,General Neuroscience ,Dentate gyrus ,Dopaminergic ,Neural Inhibition ,Sensory Gating ,Potassium channel ,Ether-A-Go-Go Potassium Channels ,Antibodies, Anti-Idiotypic ,Rats ,Acoustic Stimulation ,Acoustic Startle Reflex ,Startle reflex ,Dopamine Agonists ,Schizophrenia ,Neuroscience - Abstract
Recently, our group described the ether-à-go-go1(Eag1) voltage-gated potassium (K+) channel (Kv10.1) expression in the dopaminergic cells indicating that these channels are part of the diversified group of ion channels related to dopaminergic neurons function. The increase of dopamine neurotransmission induces a reduction in the prepulse inhibition (PPI) of the acoustic startle reflex in rodents, which is a reliable index of sensorimotor gating deficits. The PPI response has been reported to be abnormally reduced in schizophrenia patients. The role of Eag1 K+ channels in the PPI reaction had not been revealed until now, albeit the singular distribution of Eag1 in the dentate gyrus of the hippocampus and the hippocampal regulation of the startle reflex and PPI. The aim of this work was to investigate if Eag1 blockade on hippocampus modifies the PPI-disruptive effects of apomorphine in Wistar rats. Bilateral injection of anti-Eag1 single-chain antibody into the dentate gyrus of hippocampus did not modify apomorphine-disruptive effects in the PPI response. However, Eag1 antibody completely restored the startle amplitude decrease revealed after dentate gyrus surgery. These potentially biological important phenomenon merits further investigation regarding the role of Eag1 K+ channels, mainly, on startle reflex modulation, since the physiological role of these channels remain obscure.
- Published
- 2013
37. Influence of the Eag1 potassium channel expression on survival in patients with glioblastoma multiforme
- Author
-
Martinez, Ramon, Rohde, Veit, Schell, Julian, Martin, Sabine, and Pardo, Luis
- Subjects
ddc: 610 ,glioblastoma ,610 Medical sciences ,Medicine ,Eag1 ,survival - Abstract
Objective: Eag1, a voltage-gated potassium channel is expressed in the brain, participating in the control of cell excitability. In addition, it has been implicated in malignant transformation and tumor progression in cancer cell lines and human malignancies. Eag1 function can be blocked in vitro and[for full text, please go to the a.m. URL], 64. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC)
- Published
- 2013
38. Impact of the Eag1 potassium channel expression levels on survival in patients with brain metastases
- Author
-
Martinez, R, Rohde, V, Schell, J, Martin, S, and Pardo, L
- Subjects
ddc: 610 ,metastasis ,610 Medical sciences ,Medicine ,Eag1 ,survival - Abstract
Objective: Eag1, a voltage-gated potassium channel is expressed in the brain and participates in the control of cell excitability. In addition, it has been implicated in malignant transformation and progression in cancer cell lines and human malignancies. Eag1 function can be blocked in vitro and in[for full text, please go to the a.m. URL], 64. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC)
- Published
- 2013
- Full Text
- View/download PDF
39. Influence of the Eag1 potassium channel expression on survival in patients with glioblastoma multiforme
- Author
-
Martinez, R, Rohde, V, Schell, J, Martin, S, Pardo, L, Martinez, R, Rohde, V, Schell, J, Martin, S, and Pardo, L
- Published
- 2013
40. Astemizole inhibits cell proliferation in human prostate tumorigenic cells expressing ether à-go-go-1 potassium channels.
- Author
-
Bernal-Ramos G, Hernández-Gallegos E, Vera E, Chávez-López MG, Zúñiga-García V, Sánchez-Pérez Y, Garrido E, and Camacho J
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Movement drug effects, Ether-A-Go-Go Potassium Channels genetics, Humans, Immunohistochemistry, Male, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, Astemizole pharmacology, Cell Proliferation drug effects, Ether-A-Go-Go Potassium Channels metabolism, Gene Expression Regulation, Neoplastic drug effects
- Abstract
Prostate cancer (PC) is the main cause of cancer mortality in men worldwide. Therefore, novel treatments for PC are needed. Ether à-go-go-1 (Eag1) potassium channels display oncogenic properties, and have been suggested as early tumor markers and therapeutic targets for different cancers. These channels are overexpressed in many human tumors including PC. Astemizole targets several molecules involved in cancer including Eag1 channels, histamine receptors and ABC transporters. Here we studied Eag1 mRNA expression and protein levels in the non-tumorigenic and non-invasive human prostate RWPE-1 cell line, and in the tumorigenic and highly invasive human prostate WPE1-NB26 cell lines. The effect of astemizole on cell proliferation and apoptosis was also studied. The human prostate cell lines RWPE-1 and WPE1-NB26 were cultured following the provider´s instructions. Eag1 mRNA expression and protein levels were studied by real time RT-PCR and immunocytochemistry, respectively. Cell proliferation and apoptosis were studied by a fluorescence AlamarBlue® assay and flow cytometry, respectively. No difference in Eag1 mRNA expression was observed between the cell lines. However, high Eag1 protein levels were observed in the invasive WPE1-NB26 cells, in contrast to the weak protein expression in RWPE-1 cells. Accordingly, astemizole decreased cell proliferation at nanomolar concentrations only in the invasive WPE1-NB26 cells. Our results suggest that astemizole may have clinical relevance for prostate cancer treatment in patients with high Eag1 protein levels.
- Published
- 2017
- Full Text
- View/download PDF
41. Characterization of Eag1 Channel Lateral Mobility in Rat Hippocampal Cultures by Single-Particle-Tracking with Quantum Dots
- Author
-
David Gomez-Varela, Ralf B. Nehring, Stephan Schäfer, Manuela Schmidt, Luis A. Pardo, Walter Stühmer, Hiroshi Kawabe, Tobias Kohl, Maria E. Rubio, and Koch, Karl-Wilhelm
- Subjects
Fluorescent Antibody Technique ,lcsh:Medicine ,In Vitro Techniques ,Hippocampal formation ,Hippocampus ,Synapse ,03 medical and health sciences ,0302 clinical medicine ,Quantum Dots ,Biochemistry/Cell Signaling and Trafficking Structures ,Neuroscience/Neuronal Signaling Mechanisms ,Animals ,Eag1 ,Channel ,Lateral Mobility ,Rat ,Hippocampal Cultures ,Single-Particle-Tracking ,lcsh:Science ,Cytoskeleton ,Cells, Cultured ,Ion channel ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Voltage-gated ion channel ,Chemistry ,lcsh:R ,Actins ,Ether-A-Go-Go Potassium Channels ,Rats ,Microscopy, Electron ,Quantum dot ,Biophysics ,lcsh:Q ,Cell Biology/Nuclear Structure and Function ,030217 neurology & neurosurgery ,Intracellular ,Research Article - Abstract
Voltage-gated ion channels are main players involved in fast synaptic events. However, only slow intracellular mechanisms have so far been described for controlling their localization as real-time visualization of endogenous voltage-gated channels at high temporal and spatial resolution has not been achieved yet. Using a specific extracellular antibody and quantum dots we reveal and characterize lateral mobility as a faster mechanism to dynamically control the number of endogenous ethera- go-go (Eag)1 ion channels inside synapses. We visualize Eag1 entering and leaving synapses by lateral diffusion in the plasma membrane of rat hippocampal neurons. Mathematical analysis of their trajectories revealed how the motion of Eag1 gets restricted when the channels diffuse into the synapse, suggesting molecular interactions between Eag1 and synaptic components. In contrast, Eag1 channels switch to Brownian movement when they exit synapses and diffuse into extrasynaptic membranes. Furthermore, we demonstrate that the mobility of Eag1 channels is specifically regulated inside synapses by actin filaments, microtubules and electrical activity. In summary, using single-particle-tracking techniques with quantum dots nanocrystals, our study shows for the first time the lateral diffusion of an endogenous voltage-gated ion channel in neurons. The location-dependent constraints imposed by cytoskeletal elements together with the regulatory role of electrical activity strongly suggest a pivotal role for the mobility of voltage-gated ion channels in synaptic activity. peerReviewed
- Published
- 2010
- Full Text
- View/download PDF
42. Screening for Non-Pore-Binding Modulators of EAG K+ Channels.
- Author
-
Fernandes AS, Morais-Cabral JH, and Harley CA
- Subjects
- Animals, Binding Sites, Ether-A-Go-Go Potassium Channels chemistry, Humans, Mice, Protein Domains drug effects, Protein Structure, Tertiary, Drug Evaluation, Preclinical methods, Ether-A-Go-Go Potassium Channels antagonists & inhibitors, High-Throughput Screening Assays methods
- Abstract
Members of the ether-à-go-go (EAG) family of voltage-gated K(+) channels are involved in several pathophysiological diseases, and there has been a great interest in screening for drugs that modulate the activity of these channels. Many drugs have been shown to bind in the pore of these channels, blocking ion flux and causing disease pathology. In this report, we present two independent screening campaigns in which we wanted to identify small molecules that bind to either the intracellular cytoplasmic amino terminal Per-Arnt-Sim (PAS) domain from the human EAG-related gene (ERG) channel or the amino or carboxy terminal globular domains from the mouse EAG1 channel, affecting their interaction. We report that in both cases, compounds were identified that showed weak, nonspecific binding. We suggest alternative routes should be pursued in future efforts to identify specific, high-affinity binders to these cytoplasmic domains., (© 2016 Society for Laboratory Automation and Screening.)
- Published
- 2016
- Full Text
- View/download PDF
43. KV10.1 K(+)-channel plasma membrane discrete domain partitioning and its functional correlation in neurons.
- Author
-
Jiménez-Garduño AM, Mitkovski M, Alexopoulos IK, Sánchez A, Stühmer W, Pardo LA, and Ortega A
- Subjects
- Animals, Blotting, Western, Cell Membrane chemistry, Cholesterol metabolism, Cytoskeleton metabolism, Detergents metabolism, Electrophysiology, Ether-A-Go-Go Potassium Channels chemistry, Ether-A-Go-Go Potassium Channels genetics, Female, HEK293 Cells, Humans, Membrane Microdomains chemistry, Mice, Mice, Inbred C57BL, Neurons cytology, Potassium Channels, Voltage-Gated chemistry, Potassium Channels, Voltage-Gated genetics, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Cell Membrane metabolism, Ether-A-Go-Go Potassium Channels metabolism, Membrane Microdomains metabolism, Neurons metabolism, Potassium Channels, Voltage-Gated metabolism
- Abstract
KV10.1 potassium channels are implicated in a variety of cellular processes including cell proliferation and tumour progression. Their expression in over 70% of human tumours makes them an attractive diagnostic and therapeutic target. Although their physiological role in the central nervous system is not yet fully understood, advances in their precise cell localization will contribute to the understanding of their interactions and function. We have determined the plasma membrane (PM) distribution of the KV10.1 protein in an enriched mouse brain PM fraction and its association with cholesterol- and sphingolipid-rich domains. We show that the KV10.1 channel has two different populations in a 3:2 ratio, one associated to and another excluded from Detergent Resistant Membranes (DRMs). This distribution of KV10.1 in isolated PM is cholesterol- and cytoskeleton-dependent since alteration of those factors changes the relationship to 1:4. In transfected HEK-293 cells with a mutant unable to bind Ca(2+)/CaM to KV10.1 protein, Kv10.1 distribution in DRM/non-DRM is 1:4. Mean current density was doubled in the cholesterol-depleted cells, without any noticeable effects on other parameters. These results demonstrate that recruitment of the KV10.1 channel to the DRM fractions involves its functional regulation., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
44. Expression of Eag1 K+ channel and ErbBs in human pituitary adenomas: cytoskeleton arrangement patterns in cultured cells.
- Author
-
del Pliego MG, Aguirre-Benítez E, Paisano-Cerón K, Valdovinos-Ramírez I, Rangel-Morales C, Rodríguez-Mata V, Solano-Agama C, Martín-Tapia D, de la Vega MT, Saldoval-Balanzario M, Camacho J, and Mendoza-Garrido ME
- Subjects
- Adenoma pathology, Adenoma surgery, Adult, Aged, Cells, Cultured, Cytoskeleton pathology, Female, Humans, Male, Middle Aged, Pituitary Neoplasms pathology, Pituitary Neoplasms surgery, Adenoma metabolism, Cytoskeleton metabolism, ErbB Receptors metabolism, Ether-A-Go-Go Potassium Channels metabolism, Pituitary Neoplasms metabolism, Receptor, ErbB-2 metabolism
- Abstract
Pituitary adenomas can invade surrounded tissue, but the mechanism remains elusive. Ether à go-go-1 (Eag1) potassium channel and epidermal growth factor receptors (ErbB1 and ErbB2) have been associated to invasive phenotypes or poor prognosis in cancer patients. However, cells arrange their cytoskeleton in order to acquire a successful migration pattern. We have studied ErbBs and Eag1 expression, and cytoskeleton arrangements in 11 human pituitary adenomas. Eag1, ErbB1 and ErbB2 expression were studied by immunochemistry in tissue and cultured cells. The cytoskeleton arrangement was analyzed in cultured cells by immunofluorescence. Normal pituitary tissue showed ErbB2 expression and Eag1 only in few cells. However, Eag1 and ErbB2 were expressed in all the tumors analyzed. ErbB1 expression was observed variable and did not show specificity for a tumor characteristic. Cultured cells from micro- and macro-adenomas clinically functional organize their cytoskeleton suggesting a mesenchymal pattern, and a round leucocyte/amoeboid pattern from invasive clinically silent adenoma. Pituitary tumors over-express EGF receptors and the ErbB2 repeated expression suggests is a characteristic of adenomas. Eag 1 was express, in different extent, and could be a therapeutic target. The cytoskeleton arrangements observed suggest that pituitary tumor cells acquire different patterns: mesenchymal, and leucocyte/amoeboid, the last observed in the invasive adenomas. Amoeboid migration pattern has been associated with high invasion capacity.
- Published
- 2013
45. Tumor cell-selective apoptosis induction through targeting of KV10.1 via bifunctional TRAIL antibody
- Author
-
Hartung, F., Stühmer, W., and Pardo, L.
- Subjects
Cancer Research ,Cell Survival ,Recombinant Fusion Proteins ,Gene Expression ,Antineoplastic Agents ,Apoptosis ,lcsh:RC254-282 ,TNF-Related Apoptosis-Inducing Ligand ,Cell Line, Tumor ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,Humans ,Cycloheximide ,Eag1 ,Research ,Cell Cycle ,scFv62-TRAIL ,KV10.1 ,Bystander Effect ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Ether-A-Go-Go Potassium Channels ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Oncology ,Molecular Medicine ,Single-Chain Antibodies - Abstract
Background The search for strategies to target ion channels for therapeutic applications has become of increasing interest. Especially, the potassium channel KV10.1 (Ether-á-go-go) is attractive as target since this surface protein is virtually not detected in normal tissue outside the central nervous system, but is expressed in approximately 70% of tumors from different origins. Methods We designed a single-chain antibody against an extracellular region of KV10.1 (scFv62) and fused it to the human soluble TRAIL. The KV10.1-specific scFv62 antibody -TRAIL fusion protein was expressed in CHO-K1 cells, purified by chromatography and tested for biological activity. Results Prostate cancer cells, either positive or negative for KV10.1 were treated with the purified construct. After sensitization with cytotoxic drugs, scFv62-TRAIL induced apoptosis only in KV10.1-positive cancer cells, but not in non-tumor cells, nor in tumor cells lacking KV10.1 expression. In co-cultures with KV10.1-positive cancer cells the fusion protein also induced apoptosis in bystander KV10.1-negative cancer cells, while normal prostate epithelial cells were not affected when present as bystander. Conclusions KV10.1 represents a novel therapeutic target for cancer. We could design a strategy that selectively kills tumor cells based on a KV10.1-specific antibody.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.