Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 64
1.
Epilepsy Behav ; 106: 106894, 2020 05.
Article En | MEDLINE | ID: mdl-32222671

Voltage-gated potassium (Kv) channels are widely expressed in the central and peripheral nervous system and are crucial mediators of neuronal excitability. Importantly, these channels also actively participate in cellular and molecular signaling pathways that regulate the life and death processes of neurons. The current study used a kainic acid (KA)-induced temporal lobe epilepsy model to examine the role of the Kv10.2 gene in status epilepticus (SE). Lentiviral plasmids containing the coding sequence region of the KCNH5 gene (LV-KCNH5) were injected into the CA3 subarea of the right dorsal hippocampus within 24 h in post-SE rats to rescue Kv10.2 protein expression. Open-field and elevated plus maze test results indicated that anxiety-like behavior was ameliorated in the KA + LV-KCNH5 group rats compared with the SE group rats, and working memory was improved in the Y-maze test. However, the spatial reference memory of the LV-KCNH5 group rats did not improve in the Morris water maze test, and no difference was found in the light-dark transition box test. The results of this study indicate that Kv10.2 protein may play an important role in epilepsy, providing new potential therapeutic directions and drug targets for epilepsy treatment.


Cognition/physiology , Emotions/physiology , Ether-A-Go-Go Potassium Channels/biosynthesis , Kainic Acid/toxicity , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Animals , Cognition/drug effects , Emotions/drug effects , Ether-A-Go-Go Potassium Channels/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Lentivirus/genetics , Male , Maze Learning/drug effects , Maze Learning/physiology , Rats , Spatial Memory/drug effects , Spatial Memory/physiology , Status Epilepticus/genetics
2.
Neuroreport ; 30(9): 637-644, 2019 06 12.
Article En | MEDLINE | ID: mdl-31008829

Ion channels play as a pivotal role in hypertension in the processes of maintenance of vascular tone and sympathetic excitement of hypertension. The Kv10.2 channel (encoded by the Kcnh5 gene) belongs to the EAG voltage-gated superfamily. It is distributed widely in the brain, such as the hippocampus, the cortex, and the olfactory bulb. To date, the expression of Kv10.2 in central nervous system nuclei that regulates cardiovascular function and its inter-relationship with hypertension are still unclear. Here, electric foot-shock stressors with noise were used to establish the stress-induced hypertensive (SIH) rat model. The expression of Kv10.2 in the rostral ventrolateral medulla, the nucleus tractus solitarius, and the paraventricular nucleus (PVN) was examined by immunohistochemical staining and western blots. The following results were obtained: (a) the expression level of Kv10.2 was increased obviously in the paraventricular nucleus of SIH rats, whereas no significant difference was found in the rostral ventrolateral medulla and the nucleus tractus solitarius. (b) Kv10.2 was located in neurons. (c) Vesicular glutamate transporter 1 as a protein mark of glutamate neurons was increased in the paraventricular nucleus of the SIH group. (d) The expression of vesicular glutamate transporter 1 protein in neurons was significantly decreased when the Kcnh5 gene was knocked down by small interfering RNA in vitro. These findings indicate that the changes in Kv10.2 may be related to SIH, which may provide a potential avenue for further investigation of SIH.


Brain/metabolism , Ether-A-Go-Go Potassium Channels/biosynthesis , Hypertension/metabolism , Psychological Distress , Animals , Hypertension/etiology , Male , Rats , Rats, Sprague-Dawley
3.
J Gen Physiol ; 150(8): 1189-1201, 2018 08 06.
Article En | MEDLINE | ID: mdl-29941431

The human ether-a-go-go-related gene (hERG) encodes a voltage-gated potassium channel that controls repolarization of cardiac action potentials. Accumulating evidence suggests that most disease-related hERG mutations reduce the function of the channel by disrupting protein biogenesis of the channel in the endoplasmic reticulum (ER). However, the molecular mechanism underlying the biogenesis of ERG K+ channels is largely unknown. By forward genetic screening, we identified an ER-located chaperone CNX-1, the worm homologue of mammalian chaperone Calnexin, as a critical regulator for the protein biogenesis of UNC-103, the ERG-type K+ channel in Caenorhabditis elegans Loss-of-function mutations of cnx-1 decreased the protein level and current density of the UNC-103 K+ channel and suppressed the behavioral defects caused by a gain-of-function mutation in unc-103 Moreover, CNX-1 facilitated tetrameric assembly of UNC-103 channel subunits in a liposome-assisted cell-free translation system. Further studies showed that CNX-1 act in parallel to DNJ-1, another ER-located chaperone known to regulate maturation of UNC-103 channels, on controlling the protein biogenesis of UNC-103. Importantly, Calnexin interacted with hERG proteins in the ER in HEK293T cells. Deletion of calnexin reduced the expression and current densities of endogenous hERG K+ channels in SH-SY5Y cells. Collectively, we reveal an evolutionarily conserved chaperone CNX-1/Calnexin controlling the biogenesis of ERG-type K+ channels.


Caenorhabditis elegans Proteins/physiology , Calcium-Binding Proteins/physiology , Endoplasmic Reticulum/metabolism , Ether-A-Go-Go Potassium Channels/biosynthesis , HSP40 Heat-Shock Proteins/physiology , Potassium Channels/biosynthesis , Amino Acid Sequence , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Calcium-Binding Proteins/genetics , Calnexin/metabolism , HEK293 Cells , HSP40 Heat-Shock Proteins/genetics , Humans , Mice
4.
Oncotarget ; 7(25): 37436-37455, 2016 Jun 21.
Article En | MEDLINE | ID: mdl-27224923

Transforming growth factor ß (TGFß)-mediated anti-proliferative and differentiating effects promote neuronal differentiation during embryonic central nervous system development. TGFß downstream signals, composed of activated SMAD2/3, SMAD4 and a FOXO family member, promote the expression of cyclin-dependent kinase inhibitor Cdkn1a. In early CNS development, IGF1/PI3K signaling and the transcription factor FOXG1 inhibit FOXO- and TGFß-mediated Cdkn1a transcription. FOXG1 prevents cell cycle exit by binding to the SMAD/FOXO-protein complex. In this study we provide further details on the FOXG1/FOXO/SMAD transcription factor network. We identified ligands of the TGFß- and IGF-family, Foxo1, Foxo3 and Kcnh3 as novel FOXG1-target genes during telencephalic development and showed that FOXG1 interferes with Foxo1 and Tgfß transcription. Our data specify that FOXO1 activates Cdkn1a transcription. This process is under control of the IGF1-pathway, as Cdkn1a transcription increases when IGF1-signaling is pharmacologically inhibited. However, overexpression of CDKN1A and knockdown of Foxo1 and Foxo3 is not sufficient for neuronal differentiation, which is probably instructed by TGFß-signaling. In mature neurons, FOXG1 activates transcription of the seizure-related Kcnh3, which might be a FOXG1-target gene involved in the FOXG1 syndrome pathology.


Ether-A-Go-Go Potassium Channels/biosynthesis , Forkhead Transcription Factors/metabolism , Neurons/metabolism , Smad Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/cytology , Signal Transduction , Transfection
5.
Circ Arrhythm Electrophysiol ; 9(4): e003439, 2016 Apr.
Article En | MEDLINE | ID: mdl-27071825

BACKGROUND: Ventricular arrhythmias as a result of unintentional blockade of the Kv11.1 (hERG [human ether-à-go-go-related gene]) channel are a major safety concern in drug development. In past years, several highly prescribed drugs have been withdrawn for their ability to cause such proarrhythmia. Here, we investigated whether the proarrhythmic risk of existing drugs could be reduced by Kv11.1 allosteric modulators. METHODS AND RESULTS: Using [(3)H]dofetilide-binding assays with membranes of human Kv11.1-expressing human embryonic kidney 293 cells, 2 existing compounds (VU0405601 and ML-T531) and a newly synthesized compound (LUF7244) were found to be negative allosteric modulators of dofetilide binding to the Kv11.1 channel, with LUF7244 showing the strongest effect at 10 µmol/L. The Kv11.1 affinities of typical blockers (ie, dofetilide, astemizole, sertindole, and cisapride) were significantly decreased by LUF7244. Treatment of confluent neonatal rat ventricular myocyte (NRVM) monolayers with astemizole or sertindole caused heterogeneous prolongation of action potential duration and a high incidence of early afterdepolarizations on 1-Hz electric point stimulation, occasionally leading to unstable, self-terminating tachyarrhythmias. Pretreatment of NRVMs with LUF7244 prevented these proarrhythmic effects. NRVM monolayers treated with LUF7244 alone displayed electrophysiological properties indistinguishable from those of untreated NRVM cultures. Prolonged exposure of NRVMs to LUF7244 or LUF7244 plus astemizole did not affect their viability, excitability, and contractility as assessed by molecular, immunological, and electrophysiological assays. CONCLUSIONS: Allosteric modulation of the Kv11.1 channel efficiently suppresses drug-induced ventricular arrhythmias in vitro by preventing potentially arrhythmogenic changes in action potential characteristics, raising the possibility to resume the clinical use of unintended Kv11.1 blockers via pharmacological combination therapy.


Allosteric Regulation , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Regulation, Developmental , RNA/genetics , Tachycardia, Ventricular/genetics , Animals , Animals, Newborn , Anti-Arrhythmia Agents/toxicity , Cells, Cultured , Disease Models, Animal , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/biosynthesis , Ether-A-Go-Go Potassium Channels/drug effects , Humans , Immunohistochemistry , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tachycardia, Ventricular/chemically induced , Tachycardia, Ventricular/metabolism
6.
BMC Cancer ; 15: 839, 2015 Nov 03.
Article En | MEDLINE | ID: mdl-26530050

BACKGROUND: Kv10.1, a voltage-gated potassium channel only detected in the healthy brain, was found to be aberrantly expressed in extracerebral cancers. Investigations of Kv10.1 in brain metastasis and glioblastoma multiforme (GBM) are lacking. METHODS: We analyzed the expression of Kv10.1 by immunohistochemistry in these brain tumors (75 metastasis from different primary tumors, 71 GBM patients) and the influence of a therapy with tricyclic antidepressants (which are Kv10.1 blockers) on survival. We also investigated Kv10.1 expression in the corresponding primary carcinomas of metastases patients. RESULTS: We observed positive Kv10.1 expression in 85.3 % of the brain metastases and in 77.5 % of GBMs. Patients with brain metastases, showing low Kv10.1 expression, had a significantly longer overall survival compared to those patients with high Kv10.1 expression. Metastases patients displaying low Kv10.1 expression and also receiving tricyclic antidepressants showed a significantly longer median overall survival as compared to untreated patients. CONCLUSIONS: Our data show that Kv10.1 is not only highly expressed in malignant tumors outside CNS, but also in the most frequent cerebral cancer entities, metastasis and GBM, which remain incurable in spite of aggressive multimodal therapies. Our results extend the correlation between dismal prognosis and Kv10.1 expression to patients with brain metastases or GBMs and, moreover, they strongly suggest a role of tricyclic antidepressants for personalized therapy of brain malignancies.


Brain Neoplasms/genetics , Ether-A-Go-Go Potassium Channels/biosynthesis , Glioblastoma/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Disease-Free Survival , Ether-A-Go-Go Potassium Channels/genetics , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Male , Middle Aged , Prognosis
7.
Biomed Res Int ; 2015: 896432, 2015.
Article En | MEDLINE | ID: mdl-26339650

Because of their high incidence and mortality solid cancers are a major health problem worldwide. Although several new biomarkers and potential targets for therapy have been identified through biomolecular research in the last years, the effects on patients' outcome are still unsatisfactory. Increasing evidence indicates that hERG1 potassium channels are overexpressed in human primary cancers of different origin and several associations between hERG1 expression and clinicopathological features and/or outcome are emerging. Aberrant hERG1 expression may be exploited either for early diagnosis (especially in those cancers where it is expressed in the initial steps of tumor progression) or for therapy purposes. Indeed, hERG1 blockage impairs tumor cell growth both in vitro and in vivo in preclinical mouse model. hERG1-based tumor therapy in humans, however, encounters the major hindrance of the potential cardiotoxicity that many hERG1 blockers exert. In this review we focus on recent advances in translational research in some of the most frequent human solid cancers (breast, endometrium, ovary, pancreas, esophagus, stomach, and colorectum) that have been shown to express hERG1 and that are a major health problem.


Biomarkers, Tumor/biosynthesis , Cell Proliferation/genetics , Ether-A-Go-Go Potassium Channels/biosynthesis , Neoplasms/genetics , Animals , Biomarkers, Tumor/genetics , ERG1 Potassium Channel , Early Detection of Cancer , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/pathology , Translational Research, Biomedical
8.
Biomed Res Int ; 2015: 308475, 2015.
Article En | MEDLINE | ID: mdl-25866772

OBJECTIVE: To explore the antiprostate cancer effects of Celastrol on prostate cancer cells' proliferation, apoptosis, and cell cycle distribution, as well as the correlation to the regulation of hERG. METHODS: DU145 cells were treated with various concentrations of Celastrol (0.25-16.0 µmol/L) for 0-72 hours. MTT assay was used to evaluate the inhibition effect of Celastrol on the growth of DU145 cells. Cell apoptosis was detected through both Annexin-V FITC/PI double-labeled cytometry and Hoechst 33258. Cell cycle regulation was examined by a propidium iodide method. Western blot and RT-PCR technologies were applied to assess the expression level of hERG in DU145 cells. RESULTS: Celastrol presented striking growth inhibition and apoptosis induction potency on DU145 cells in vitro in a time- and dose-dependent manner. The IC50 value of Celastrol for 24 hours was 2.349 ± 0.213 µmol/L. Moreover, Celastrol induced DU145 cell apoptosis in a cell cycle-dependent manner, which means Celastrol could arrest DU145 cells in G0/G1 phase; accordingly, cells in S phase decreased gradually and no obvious changes were found in G2/M phase cells. Through transmission electron microscope, apoptotic bodies containing nuclear fragments were found in Celastrol-treated DU145 cells. Overexpression of hERG channel was found in DU145 cells, while Celastrol could downregulate it at both protein and mRNA level in a dose-dependent manner (P < 0.01). CONCLUSIONS: Celastrol exhibits its antiprostate cancer effects partially through the downregulation of the expression level of hERG channel in DU145 cells, suggesting that Celastrol may be a potential agent against prostate cancer with a mechanism of blocking the hERG channel.


Cell Proliferation/drug effects , Down-Regulation/drug effects , Ether-A-Go-Go Potassium Channels/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Triterpenes/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Humans , Male , Neoplasm Proteins/genetics , Pentacyclic Triterpenes , Prostatic Neoplasms
9.
Tumour Biol ; 36(8): 6149-58, 2015 Aug.
Article En | MEDLINE | ID: mdl-25783527

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.


Astemizole/administration & dosage , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Ether-A-Go-Go Potassium Channels/biosynthesis , Liver Neoplasms/genetics , Animals , Apoptosis/drug effects , Biomarkers, Tumor/biosynthesis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Diethylnitrosamine/administration & dosage , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Neoplasm Staging , Prognosis , Rats , gamma-Glutamyltransferase/biosynthesis
10.
Microb Cell Fact ; 14: 15, 2015 Feb 07.
Article En | MEDLINE | ID: mdl-25656388

The hERG potassium channel is essential for repolarization of the cardiac action potential. Due to this vital function, absence of unintended and potentially life-threatening interactions with hERG is required for approval of new drugs. The structure of hERG is therefore one of the most sought-after. To provide purified hERG for structural studies and new hERG biomimetic platforms for detection of undesirable interactions, we have developed a hERG expression platform generating unprecedented amounts of purified and functional hERG channels. Full-length hERG, with or without a C-terminally fused green fluorescent protein (GFP) His 8-tag was produced from a codon-optimized hERG cDNA in Saccharomyces cerevisiae. Both constructs complemented the high potassium requirement of a knock-out Saccharomyces cerevisiae strain, indicating correct tetramer assembly in vivo. Functionality was further demonstrated by Astemizole binding to membrane embedded hERG-GFP-His 8 with a stoichiometry corresponding to tetramer assembly. The 156 kDa hERG-GFP protein accumulated to a membrane density of 1.6%. Fluorescence size exclusion chromatography of hERG-GFP-His 8 solubilized in Fos-Choline-12 supplemented with cholesteryl-hemisuccinate and Astemizole resulted in a monodisperse elution profile demonstrating a high quality of the hERG channels. hERG-GFP-His 8 purified by Ni-affinity chromatography maintained the ability to bind Astemizole with the correct stoichiometry indicating that the native, tetrameric structure was preserved. To our knowledge this is the first reported high-yield production and purification of full length, tetrameric and functional hERG. This significant breakthrough will be paramount in obtaining hERG crystal structures, and in establishment of new high-throughput hERG drug safety screening assays.


Astemizole/metabolism , Ether-A-Go-Go Potassium Channels/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Biomass , Cell Membrane/metabolism , Chromatography, Affinity/methods , DNA, Complementary/genetics , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/isolation & purification , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histamine H1 Antagonists, Non-Sedating/metabolism , Humans , Microscopy, Fluorescence , Protein Binding , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Saccharomyces cerevisiae/genetics , Temperature , Time Factors
11.
Int J Clin Exp Pathol ; 8(11): 15093-9, 2015.
Article En | MEDLINE | ID: mdl-26823849

BACKGROUNDS: Expression of eag1 channel (Eag1) is associated with cell malignant transformation, tumor cell metastasis and poor prognosis of the patient. This study aimed at examining whether expression of the Eag1 associated with aggressive clinicopathological feature and the molecular subtype of breast cancer. MATERIALS AND METHODS: 109 patients who received breast cancer operation during January 2009 to December 2010 in Chinese-Japanese Friendship Hospital of Jilin University were recruited. We investigated the association of the Eag1 with clinicopathological features and molecular subtype of in triple negative breast cancer (TNBC) by univariate or multivariate analysis in a cross-section study. RESULTS: The positive rate of Eag1 was 18.5% higher in TNBC compared with non-triple negative breast cancer (Non-TNBC) (P = 0.012, OR = 2.83, 95% CI = 2.16-3.47). Compared with the Eag1 negative group, the expression of Eag1 was linked to the larger tumor size (P = 0.002), advanced TNM stage (P = 0.029), high proportion of positive lymph node (87.6% vs. 65%, P = 0.014) and invasive ductal carcinoma (91% vs. 75%, P = 0.046). CONCLUSIONS: The expression of Eag1 may be partially explained the aggressive behavior of TNBC in the breast cancer tissue.


Biomarkers, Tumor/analysis , Carcinoma, Ductal, Breast/pathology , Ether-A-Go-Go Potassium Channels/biosynthesis , Triple Negative Breast Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Cross-Sectional Studies , Female , Humans , Immunohistochemistry , Middle Aged
12.
Biomed Res Int ; 2015: 617316, 2015.
Article En | MEDLINE | ID: mdl-26783521

So far, the role of Ether à go-go 1 (Eag1) potassium channels in migration and invasion progression of cancers remains elusive. In the present study, the effects of Eag1 knockdown on osteosarcoma cell proliferation, growth, and apoptosis were examined. Then, we evaluated the effects of Eag1 silencing on osteosarcoma cell migration and invasion. In addition, we detected the expression of vascular endothelial growth factor (VEGF) and signal transducer and activator of transcription 3 (STAT3) in osteosarcoma cell treated with Eag1 small interfering RNAs (siRNAs). Finally, STAT3 siRNA was employed to determine the influence of downregulation of STAT3 on cell proliferation and migration. The results showed that knockdown of Eag1 significantly suppressed osteosarcoma cell proliferation and osteosarcoma xenografts growth. However, Eag1 silencing had little effect on cell apoptosis. Additionally, osteosarcoma cell adhesion, migration, and invasion were also potently attenuated. Notably, the expression levels of VEGF decreased evidently upon Eag1 siRNAs treatment, paralleled with reductions in the expression levels of STAT3. Moreover, a similar pattern was observed in osteosarcoma cell proliferation and migration suppression between STAT3 siRNA and Eag1 siRNAs groups. Our data indicated that Eag1 promotes osteosarcoma proliferation and migration, at least in part, by targeting STAT3-VEGF pathway.


Ether-A-Go-Go Potassium Channels/genetics , Osteosarcoma/genetics , STAT3 Transcription Factor/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/biosynthesis , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Osteosarcoma/pathology , Signal Transduction/genetics , Xenograft Model Antitumor Assays
13.
Biomed Res Int ; 2014: 345678, 2014.
Article En | MEDLINE | ID: mdl-25136578

The ether à go-go1 (Eag1) channel is overexpressed in a variety of cancers. However, the expression and function of Eag1 in liposarcoma are poorly understood. In the present study, the mRNA expression of Eag1 in different adipose tissue samples was examined by real-time PCR. Then, the protein expression of Eag1 in 131 different adipose tissues from 109 patients was detected by immunohistochemistry. Next, the associations between Eag1 expression and clinicopathological features of liposarcoma were analyzed. In addition, the effects of Eag1 on liposarcoma cell proliferation and cycle were evaluated by CCK-8, colony formation, xenograft mouse model, and flow cytometry, respectively. Finally, the activation of p38 mitogen-activated protein kinase (MAPK) was detected by Western blot analysis to explain the detailed mechanisms of oncogenic potential of Eag1 in liposarcoma. It was found that Eag1 was aberrantly expressed in over 67% liposarcomas, with a higher frequency than in lipoma, hyperplasia, inflammation, and normal adipose tissues. However, Eag1 expression was not correlated with clinicopathological features of liposarcoma. Eag1 inhibitor imipramine or Eag1-shRNA significantly suppressed the proliferation of liposarcoma cells in vitro and in vivo, accompanying with accumulation of cells in the G1 phase. These results suggest that Eag1 plays an important role in regulating the proliferation and cell cycle of liposarcoma cells and might be a potential therapeutic target for liposarcoma.


Adipose Tissue/metabolism , Cell Proliferation , Ether-A-Go-Go Potassium Channels/biosynthesis , Gene Expression Regulation, Neoplastic , Liposarcoma/metabolism , Neoplasm Proteins/biosynthesis , Adipose Tissue/pathology , Adult , Aged , Animals , Cell Line, Tumor , Ether-A-Go-Go Potassium Channels/genetics , Female , Heterografts , Humans , Liposarcoma/genetics , Liposarcoma/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Transplantation , p38 Mitogen-Activated Protein Kinases
14.
BMJ Open ; 4(5): e005049, 2014 May 16.
Article En | MEDLINE | ID: mdl-24838727

OBJECTIVES: To explore the expression patterns of Eag1 (ether á go-go 1) and HIF-1α (hypoxia-inducible factor 1α) in a cohort of patients with breast cancer. SETTING: Department of general surgery in an upper first-class hospital in Xi'an, China. PARTICIPANTS: A total of 112 female Han Chinese patients with a diagnosis of invasive ductal carcinoma were included. Patients with main internal diseases, such as cardiovascular, endocrine, gastroenterological, haematological, infectious diseases, etc, were excluded. PRIMARY AND SECONDARY OUTCOME MEASURES: Expression profiles of Eag1 and HIF-1α. RESULTS: Eag1 and HIF-1α were overexpressed in the tumour tissues compared with the pair-matched control tissues, p=0.002 and <0.001, respectively. The expression of Eag1 and HIF-1α was negatively correlated with tumour size, p=0.032 and p=0.025, respectively, and lymph node status (p=0.040, p=0.032, respectively). The coexpression of Eag1 and HIF-1α was correlated with tumour size (p=0.012), lymph node status (p=0.027) and tumour stage (p=0.036). HIF-1α has a strong correlation with hEag1 expression (κ=0.731, p<0.001). CONCLUSIONS: HIF-1α expression has a strong correlation with hEag1 expression. We are the first to attempt to explore the correlation at the population level.


Breast Neoplasms/metabolism , Ether-A-Go-Go Potassium Channels/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/diagnosis , Female , Humans , Immunohistochemistry , Middle Aged , Prognosis , Young Adult
15.
Diagn Mol Pathol ; 22(4): 215-21, 2013 Dec.
Article En | MEDLINE | ID: mdl-24193004

Cancer molecular investigation revealed a huge molecular heterogeneity between different types of cancers as well as among cancer patients affected by the same cancer type. This implies the necessity of a personalized approach for cancer diagnosis and therapy, on the basis of the development of standardized protocols to facilitate the application of molecular techniques in the clinical decision-making process. Ion channels encoding genes are acquiring increasing relevance in oncological translational studies, representing new candidates for molecular diagnostic and therapeutic purposes. Hence, the development of molecular protocols for the quantification of ion channels encoding genes in tumor specimens may have relevance for diagnostic and prognostic investigation. Two main hindrances must be overcome for these purposes: the use of formalin-fixed and paraffin-embedded samples for gene expression analysis and the physiological expression of ion channels in excitable cells, potentially present in the tumor sample. We here propose a method for hERG1 gene quantification in colorectal cancer samples in both cryopreserved and formalin-fixed and paraffin-embedded samples. An analytical method was developed to estimate hERG1 gene expression exclusively in epithelial cancer cells. Indeed, we found that the hERG1 gene was expressed at significant levels by myofibroblasts present in the tumor stroma. This method was based on the normalization on a smooth muscle-myofibroblast-specific gene, MYH11, with no need of microdissection. By applying this method, hERG1 expression turned out to correlate with VEGF-A expression, confirming previous immunohistochemical data.


Colorectal Neoplasms/pathology , Ether-A-Go-Go Potassium Channels/biosynthesis , Gene Expression Profiling/methods , Gene Expression , Pathology, Molecular/methods , Ether-A-Go-Go Potassium Channels/genetics , Humans , Specimen Handling/methods , Tissue Fixation/methods , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
16.
Mol Biol Cell ; 24(24): 3787-804, 2013 Dec.
Article En | MEDLINE | ID: mdl-24152733

Membrane trafficking in concert with the peripheral quality control machinery plays a critical role in preserving plasma membrane (PM) protein homeostasis. Unfortunately, the peripheral quality control may also dispose of partially or transiently unfolded polypeptides and thereby contribute to the loss-of-expression phenotype of conformational diseases. Defective functional PM expression of the human ether-a-go-go-related gene (hERG) K(+) channel leads to the prolongation of the ventricular action potential that causes long QT syndrome 2 (LQT2), with increased propensity for arrhythmia and sudden cardiac arrest. LQT2 syndrome is attributed to channel biosynthetic processing defects due to mutation, drug-induced misfolding, or direct channel blockade. Here we provide evidence that a peripheral quality control mechanism can contribute to development of the LQT2 syndrome. We show that PM hERG structural and metabolic stability is compromised by the reduction of extracellular or intracellular K(+) concentration. Cardiac glycoside-induced intracellular K(+) depletion conformationally impairs the complex-glycosylated channel, which provokes chaperone- and C-terminal Hsp70-interacting protein-dependent polyubiquitination, accelerated internalization, and endosomal sorting complex required for transport-dependent lysosomal degradation. A similar mechanism contributes to the down-regulation of PM hERG harboring LQT2 missense mutations, with incomplete secretion defect. These results suggest that PM quality control plays a determining role in the loss-of-expression phenotype of hERG in certain hereditary and acquired LTQ2 syndromes.


Cell Membrane/genetics , Ether-A-Go-Go Potassium Channels/genetics , Long QT Syndrome/genetics , Protein Transport/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cardiac Glycosides/pharmacology , Cardiotonic Agents/pharmacology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Digoxin/pharmacology , Down-Regulation , Endosomal Sorting Complexes Required for Transport/metabolism , Enzyme Inhibitors/pharmacology , Ether-A-Go-Go Potassium Channels/biosynthesis , HEK293 Cells , HeLa Cells , Heart/physiology , Humans , Ouabain/pharmacology , Patch-Clamp Techniques , Potassium/metabolism , Protein Folding , RNA Interference , RNA, Small Interfering , Ubiquitination/genetics
17.
Toxicol Appl Pharmacol ; 272(1): 245-55, 2013 Oct 01.
Article En | MEDLINE | ID: mdl-23707608

Tyrosine kinase inhibitors (TKi) have greatly improved the treatment and prognosis of multiple cancer types. However, unexpected cardiotoxicity has arisen in a subset of patients treated with these agents that was not wholly predicted by pre-clinical testing, which centers around animal toxicity studies and inhibition of the human Ether-à-go-go-Related Gene (hERG) channel. Therefore, we sought to determine whether a multi-parameter test panel assessing the effect of drug treatment on cellular, molecular, and electrophysiological endpoints could accurately predict cardiotoxicity. We examined how 4 FDA-approved TKi agents impacted cell viability, apoptosis, reactive oxygen species (ROS) generation, metabolic status, impedance, and ion channel function in human cardiomyocytes. The 3 drugs clinically associated with severe cardiac adverse events (crizotinib, sunitinib, nilotinib) all proved to be cardiotoxic in our in vitro tests while the relatively cardiac-safe drug erlotinib showed only minor changes in cardiac cell health. Crizotinib, an ALK/MET inhibitor, led to increased ROS production, caspase activation, cholesterol accumulation, disruption in cardiac cell beat rate, and blockage of ion channels. The multi-targeted TKi sunitinib showed decreased cardiomyocyte viability, AMPK inhibition, increased lipid accumulation, disrupted beat pattern, and hERG block. Nilotinib, a second generation Bcr-Abl inhibitor, led to increased ROS generation, caspase activation, hERG block, and an arrhythmic beat pattern. Thus, each drug showed a unique toxicity profile that may reflect the multiple mechanisms leading to cardiotoxicity. This study demonstrates that a multi-parameter approach can provide a robust characterization of drug-induced cardiomyocyte damage that can be leveraged to improve drug safety during early phase development.


Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/toxicity , Protein-Tyrosine Kinases/antagonists & inhibitors , Caspase 3/metabolism , Caspase 7/metabolism , Cell Survival/drug effects , Cells, Cultured , Cholesterol/metabolism , Crizotinib , ERG1 Potassium Channel , Enzyme Activation/drug effects , Erlotinib Hydrochloride , Ether-A-Go-Go Potassium Channels/biosynthesis , Ether-A-Go-Go Potassium Channels/genetics , Humans , Indoles/toxicity , Ion Channels/drug effects , Lipids/biosynthesis , Myocytes, Cardiac/ultrastructure , Patch-Clamp Techniques , Pluripotent Stem Cells/drug effects , Pyrazoles/toxicity , Pyridines/toxicity , Pyrimidines/toxicity , Pyrroles/toxicity , Quinazolines/toxicity , RNA/biosynthesis , RNA/isolation & purification , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Sunitinib
18.
J Biol Chem ; 288(21): 15075-84, 2013 May 24.
Article En | MEDLINE | ID: mdl-23589291

The hERG (human ether-a-go-go-related gene) encodes the α subunit of the rapidly activating delayed rectifier potassium channel (IKr). Dysfunction of hERG channels due to mutations or certain medications causes long QT syndrome, which can lead to fatal ventricular arrhythmias or sudden death. Although the abundance of hERG in the plasma membrane is a key determinant of hERG functionality, the mechanisms underlying its regulation are not well understood. In the present study, we demonstrated that overexpression of the stress-responsive serum- and glucocorticoid-inducible kinase (SGK) isoforms SGK1 and SGK3 increased the current and expression level of the membrane-localized mature proteins of hERG channels stably expressed in HEK 293 (hERG-HEK) cells. Furthermore, the synthetic glucocorticoid, dexamethasone, increased the current and abundance of mature ERG proteins in both hERG-HEK cells and neonatal cardiac myocytes through the enhancement of SGK1 but not SGK3 expression. We have previously shown that mature hERG channels are degraded by ubiquitin ligase Nedd4-2 via enhanced channel ubiquitination. Here, we showed that SGK1 or SGK3 overexpression increased Nedd4-2 phosphorylation, which is known to inhibit Nedd4-2 activity. Nonetheless, disruption of the Nedd4-2 binding site in hERG channels did not eliminate the SGK-induced increase in hERG expression. Additional disruption of Rab11 proteins led to a complete elimination of SGK-mediated increase in hERG expression. These results show that SGK enhances the expression level of mature hERG channels by inhibiting Nedd4-2 as well as by promoting Rab11-mediated hERG recycling.


Endosomal Sorting Complexes Required for Transport/biosynthesis , Ether-A-Go-Go Potassium Channels/biosynthesis , Gene Expression Regulation/physiology , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/biosynthesis , rab GTP-Binding Proteins/metabolism , Animals , Binding Sites , Dexamethasone/pharmacology , ERG1 Potassium Channel , Endosomal Sorting Complexes Required for Transport/genetics , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nedd4 Ubiquitin Protein Ligases , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Rats , Ubiquitin-Protein Ligases/genetics , rab GTP-Binding Proteins/genetics
19.
Melanoma Res ; 23(3): 185-90, 2013 Jun.
Article En | MEDLINE | ID: mdl-23524483

Cutaneous melanoma represents the main cause of death among skin cancers. The thickness of the lesion at diagnosis is one of the most important prognostic indicators for survival, which is good for thin melanomas (≤1 mm) and worsens as thickness increases. Nevertheless, it is not rare to observe disease progression of thin melanomas or, conversely, a good outcome for those melanomas considered to be at high risk, according to the classical prognostic criteria. In the present paper, we analysed for the first time the expression of the hERG1 protein, a potassium channel frequently overexpressed and misexpressed in cancers, in cutaneous melanocytic lesions. The analysis was carried out on archival samples relative to (a) typical melanocytic nevi, (b) atypical melanocytic nevi, (c) thin (<1 mm) melanomas from patients who survived at least 10 years after surgery, (d) thick (>4 mm) melanomas from patients who died for melanoma and (e) melanoma metastases. Samples were analysed by immunohistochemistry using an hERG1-specific antibody. We showed that primary cutaneous melanomas with a thickness greater than 4 mm as well as metastatic melanoma lesions are characterized by a high level of hERG1 expression. Conversely, thin melanomas and benign melanocytic lesions (e.g. typical and atypical melanocytic nevi) express hERG1 at significantly lower levels. Although still preliminary, the data presented here enable us to consider hERG1 as a novel candidate biomarker for aggressive melanoma.


Ether-A-Go-Go Potassium Channels/biosynthesis , Melanoma/metabolism , Skin Neoplasms/metabolism , Disease Progression , ERG1 Potassium Channel , Humans , Immunohistochemistry , Melanoma/diagnosis , Melanoma/pathology , Prognosis , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology
20.
Acta Pharmacol Sin ; 34(3): 329-35, 2013 Mar.
Article En | MEDLINE | ID: mdl-23459091

To date, research on the human ether-a-go-go related gene (hERG) has focused on this potassium channel's role in cardiac repolarization and Long QT Syndrome (LQTS). However, growing evidence implicates hERG in a diversity of physiologic and pathological processes. Here we discuss these other functions of hERG, particularly their impact on diseases beyond cardiac arrhythmia.


Ether-A-Go-Go Potassium Channels/physiology , Gene Expression , Long QT Syndrome/metabolism , Neoplasms/metabolism , Animals , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/biosynthesis , Ether-A-Go-Go Potassium Channels/genetics , Humans , Long QT Syndrome/genetics , Neoplasms/genetics , Organ Specificity
...