Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Article in English | MEDLINE | ID: mdl-34716268

ABSTRACT

The human ERG (hERG) K+ channel has a crucial function in cardiac repolarization, and mutations or channel block can give rise to long QT syndrome and catastrophic ventricular arrhythmias. The cytosolic assembly formed by the Per-Arnt-Sim (PAS) and cyclic nucleotide binding homology (CNBh) domains is the defining structural feature of hERG and related KCNH channels. However, the molecular role of these two domains in channel gating remains unclear. We have previously shown that single-chain variable fragment (scFv) antibodies can modulate hERG function by binding to the PAS domain. Here, we mapped the scFv2.12 epitope to a site overlapping with the PAS/CNBh domain interface using NMR spectroscopy and mutagenesis and show that scFv binding in vitro and in the cell is incompatible with the PAS interaction with CNBh. By generating a fluorescently labeled scFv2.12, we demonstrate that association with the full-length hERG channel is state dependent. We detect Förster resonance energy transfer (FRET) with scFv2.12 when the channel gate is open but not when it is closed. In addition, state dependence of scFv2.12 FRET signal disappears when the R56Q mutation, known to destabilize the PAS-CNBh interaction, is introduced in the channel. Altogether, these data are consistent with an extensive structural alteration of the PAS/CNBh assembly when the cytosolic gate opens, likely favoring PAS domain dissociation from the CNBh domain.


Subject(s)
ERG1 Potassium Channel/metabolism , Cyclic Nucleotide-Gated Cation Channels/metabolism , Cytosol/metabolism , ERG1 Potassium Channel/genetics , ERG1 Potassium Channel/immunology , Ether-A-Go-Go Potassium Channels/immunology , Ether-A-Go-Go Potassium Channels/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Ion Channel Gating , Long QT Syndrome/genetics , Molecular Conformation , Mutation , Protein Conformation , Protein Domains/genetics , Protein Domains/immunology , Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship
2.
Pathol Oncol Res ; 23(4): 761-767, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28070819

ABSTRACT

Ether à-go-go 1 (KCNH1, Kv10.1) (Eag1) is a voltage-gated potassium channel, which is commonly overexpressed in tested breast cancer patients. This occurrence makes it a potential molecular marker and a promising tool for breast cancer diagnosis and therapy. In order to explore protective or specific polyclonal antibodies for further research, potential linear epitopes from Eag1 were collected by sequence alignment. The sequence was synthesized and then coupled to the carrier protein keyhole limpet hemocyanin (KLH) for animal immunization. Polyclonal antibodies against Eag1 were produced and purified from the rabbit antisera. Enzyme linked immunosorbent assay (ELISA) and western blot were performed to characterize their specificities. Immunohistochemical staining was carried out on normal and cancerous breast tissue sections using the purified polyclonal Eag1-specific antibodies. The results indicate that the overexpression of Eag1 might be associated with an increased risk of progression to breast cancer (Grade 1 tissue = 57.89%;Grade 2 tissue = 92.59%;Grade 3 tissue = 100%). These results also suggest that Eag1 gene is a putative growth-promoting gene that might be involved in breast tumorigenesis and development. Eag1 might further be represented as a potential target for some human diseases treatment.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Ether-A-Go-Go Potassium Channels/immunology , Ether-A-Go-Go Potassium Channels/metabolism , Animals , Antibodies , Antibody Specificity , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Female , Humans , Immunization , Rabbits
3.
Proc Natl Acad Sci U S A ; 113(35): 9916-21, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27516548

ABSTRACT

The human human ether-à-go-go-related gene (hERG) potassium channel plays a critical role in the repolarization of the cardiac action potential. Changes in hERG channel function underlie long QT syndrome (LQTS) and are associated with cardiac arrhythmias and sudden death. A striking feature of this channel and KCNH channels in general is the presence of an N-terminal Per-Arnt-Sim (PAS) domain. In other proteins, PAS domains bind ligands and modulate effector domains. However, the PAS domains of KCNH channels are orphan receptors. We have uncovered a family of positive modulators of hERG that specifically bind to the PAS domain. We generated two single-chain variable fragments (scFvs) that recognize different epitopes on the PAS domain. Both antibodies increase the rate of deactivation but have different effects on channel activation and inactivation. Importantly, we show that both antibodies, on binding to the PAS domain, increase the total amount of current that permeates the channel during a ventricular action potential and significantly reduce the action potential duration recorded in human cardiomyocytes. Overall, these molecules constitute a previously unidentified class of positive modulators and establish that allosteric modulation of hERG channel function through ligand binding to the PAS domain can be attained.


Subject(s)
Action Potentials/drug effects , Ether-A-Go-Go Potassium Channels/physiology , Ion Channel Gating/drug effects , Single-Chain Antibodies/pharmacology , Animals , Binding Sites/genetics , Binding Sites/immunology , Cells, Cultured , Chickens , Electric Stimulation/methods , Epitopes/genetics , Epitopes/immunology , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/immunology , HEK293 Cells , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Single-Chain Antibodies/immunology
4.
Eur Biophys J ; 45(7): 721-733, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27444284

ABSTRACT

The Kv10.1 (Eag1) voltage-gated potassium channel represents a promising molecular target for novel cancer therapies or diagnostic purposes. Physiologically, it is only expressed in the brain, but it was found overexpressed in more than 70 % of tumours of diverse origin. Furthermore, as a plasma membrane protein, it is easily accessible to extracellular interventions. In this study we analysed the feasibility of the anti-Kv10.1 monoclonal antibody mAb62 to target tumour cells in vitro and in vivo and to deliver therapeutics to the tumour. Using time-domain near infrared fluorescence (NIRF) imaging in a subcutaneous MDA-MB-435S tumour model in nude mice, we showed that mAb62-Cy5.5 specifically accumulates at the tumour for at least 1 week in vivo with a maximum intensity at 48 h. Blocking experiments with an excess of unlabelled mAb62 and application of the free Cy5.5 fluorophore demonstrate specific binding to the tumour. Ex vivo NIRF imaging of whole tumours as well as NIRF imaging and microscopy of tumour slices confirmed the accumulation of the mAb62-Cy5.5 in tumours but not in brain tissue. Moreover, mAb62 was conjugated to the prodrug-activating enzyme ß-D-galactosidase (ß-gal; mAb62-ß-gal). The ß-gal activity of the mAb62-ß-gal conjugate was analysed in vitro on Kv10.1-expressing MDA-MB-435S cells in comparison to control AsPC-1 cells. We show that the mAb62-ß-gal conjugate possesses high ß-gal activity when bound to Kv10.1-expressing MDA-MB-435S cells. Moreover, using the ß-gal activatable NIRF probe DDAOG, we detected mAb62-ß-gal activity in vivo over the tumour area. In summary, we could show that the anti-Kv10.1 antibody is a promising tool for the development of novel concepts of targeted cancer therapy.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Transformation, Neoplastic , Ether-A-Go-Go Potassium Channels/immunology , Optical Imaging/methods , Animals , Carbocyanines/metabolism , Cell Line, Tumor , Ether-A-Go-Go Potassium Channels/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice
5.
J Physiol ; 594(21): 6175-6187, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27296897

ABSTRACT

KEY POINTS: Channelopathies of autoimmune origin are novel and are associated with corrected QT (QTc) prolongation and complex ventricular arrhythmias. We have recently demonstrated that anti-SSA/Ro antibodies from patients with autoimmune diseases and with QTc prolongation on the ECG target the human ether-à-go-go-related gene (HERG) K+ channel by inhibiting the corresponding current, IKr , at the pore region. Immunization of guinea-pigs with a peptide (E-pore peptide) corresponding to the extracellular loop region connecting the S5 and S6 segments of the HERG channel induces high titres of antibodies that inhibit IKr , lengthen the action potential and cause QTc prolongation on the surface ECG. In addition, anti-SSA/Ro-positive sera from patients with connective tissue diseases showed high reactivity to the E-pore peptide. The translational impact is the development of a peptide-based approach for the diagnosis and treatment of autoimmune-associated long QT syndrome. ABSTRACT: We recently demonstrated that anti-SSA/52 kDa Ro antibodies (Abs) from patients with autoimmune diseases and corrected QT (QTc) prolongation directly target and inhibit the human ether-à-go-go-related gene (HERG) K+ channel at the extracellular pore (E-pore) region, where homology with SSA/52 kDa Ro antigen was demonstrated. We tested the hypothesis that immunization of guinea-pigs with a peptide corresponding to the E-pore region (E-pore peptide) will generate pathogenic inhibitory Abs and cause QTc prolongation. Guinea-pigs were immunized with a 31-amino-acid peptide corresponding to the E-pore region of HERG. On days 10-62 after immunization, ECGs were recorded and blood was sampled for the detection of E-pore peptide Abs. Serum samples from patients with autoimmune diseases were evaluated for reactivity to E-pore peptide by enzyme-linked immunosorbent assay (ELISA), and histology was performed on hearts using Masson's Trichrome. Inhibition of the HERG channel was assessed by electrophysiology and by computational modelling of the human ventricular action potential. The ELISA results revealed the presence of high titres of E-pore peptide Abs and significant QTc prolongation after immunization. High reactivity to E-pore peptide was found using anti-SSA/Ro Ab-positive sera from patients with QTc prolongation. Histological data showed no evidence of fibrosis in immunized hearts. Simulations of simultaneous inhibition of repolarizing currents by anti-SSA/Ro Ab-positive sera showed the predominance of the HERG channel in controlling action potential duration and the QT interval. These results are the first to demonstrate that inhibitory Abs to the HERG E-pore region induce QTc prolongation in immunized guinea-pigs by targeting the HERG channel independently from fibrosis. The reactivity of anti-SSA/Ro Ab-positive sera from patients with connective tissue diseases with the E-pore peptide opens novel pharmacotherapeutic avenues in the diagnosis and management of autoimmune-associated QTc prolongation.


Subject(s)
Autoimmunity , Ether-A-Go-Go Potassium Channels/immunology , Long QT Syndrome/immunology , Animals , Antibodies/immunology , Cells, Cultured , Ether-A-Go-Go Potassium Channels/chemistry , Guinea Pigs , HEK293 Cells , Humans , Peptide Fragments/immunology
6.
Eur Biophys J ; 45(7): 709-719, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27350552

ABSTRACT

Resisting cell death is one of the hallmarks of cancer, and represents a common problem resulting in ineffective cancer therapy. To overcome resistance to apoptosis, we designed an antibody-based therapy strategy using Kv10.1 as a target. Kv10.1 is a voltage-gated potassium channel, which has been identified as a tumor marker several years ago. The agent consists of a Kv10.1-specific single-chain antibody fused to the soluble tumor necrosis factor-related apoptosis-inducing ligand (scFv62-TRAIL). We combined scFv62-TRAIL with different chemotherapeutic drugs, all of which failed to induce apoptosis when used alone. In the combination, we could overcome the resistance and selectively induce apoptosis. Among the drugs, doxorubicin showed the most promising effect. Additionally, we observed improved efficacy by pre-treating the cells with doxorubicin before scFv62-TRAIL application. Expression analysis of the TRAIL death receptors suggests a doxorubicin-induced increase in the abundance of receptors as the mechanism for sensitization. Furthermore, we confirmed the anti-tumor effect and efficacy of our combination strategy in vivo in SCID mice bearing subcutaneous tumors. In conclusion, we propose a novel strategy to overcome resistance to chemotherapy in cancer cells. Doxorubicin and scFv62-TRAIL reciprocally sensitize the cells to each other, specifically in Kv10.1-positive tumor cells.


Subject(s)
Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Ether-A-Go-Go Potassium Channels/immunology , Single-Chain Antibodies/immunology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/immunology , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Cricetinae , Cricetulus , Drug Resistance, Neoplasm/immunology , Female , Humans , Mice , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
7.
Biochem Biophys Res Commun ; 431(1): 70-5, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23277102

ABSTRACT

The human ether-a-go-go related gene (hERG) potassium channel plays a major role in the repolarization of the cardiac action potential. Inhibition of the hERG function by mutations or a wide variety of pharmaceutical compounds cause long QT syndrome and lead to potentially lethal arrhythmias. For detailed insights into the structural and biochemical background of hERG function and drug binding, the purification of recombinant protein is essential. Because the hERG channel is a challenging protein to purify, fast and easy techniques to evaluate different expression, solubilization and purification conditions are of primary importance. Here, we describe the generation of a set of 12 monoclonal antibodies against hERG. Beside their suitability in western blot, immunoprecipitation and immunostaining, these antibodies were used to establish a sandwich ELISA for the detection and relative quantification of hERG in different expression systems. Furthermore, a Fab fragment was used in fluorescence size exclusion chromatography to determine the oligomeric state of hERG after solubilization. These new tools can be used for a fast and efficient screening of expression, solubilization and purification conditions.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Enzyme-Linked Immunosorbent Assay , Ether-A-Go-Go Potassium Channels/analysis , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Chromatography, Gel/methods , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/immunology , Ether-A-Go-Go Potassium Channels/isolation & purification , HEK293 Cells , Humans , Immunoglobulin Fab Fragments , Mice
8.
Neoplasma ; 59(2): 207-15, 2012.
Article in English | MEDLINE | ID: mdl-22248279

ABSTRACT

Human ether à go-go (hEAG) potassium channels are primarily expressed in brain but also frequently overexpressed in solid tumors, which could indicate their potential value for cancer diagnosis and therapy. hEAG1, one member of the hEAG subfamily, has been shown to play a role in neoplastic process. Here we report the expression of hEAG1 in human osteosarcoma detected by a new polyclonal antibody. The full-length hEAG1 cDNA was cloned from human osteosarcoma cell line MG63 by RT-PCR and expressed in Escherichia coli as His tagged protein. The 6His-hEAG1F protein was purified by nickel agarose and used as the antigen to immunize rabbits following standard protocols. The obtained antiserum could detect hEAG1 exogenously expressed in HEK 293 cells. Furthermore, the polyclonal antibody was used to evaluate hEAG1 expression in 42 human osteosarcoma specimens and 19 osteochondromas specimens by immunohistochemistry. hEAG1 was expressed in 71.4% (30/42) osteosarcoma, and 15.8% (3/19) osteochondromas. Moreover, statistical analysis revealed that hEAG1 expression was not dependent on age, sex, site, histology, grade and type in the osteosarcoma specimens. Our data provide evidence that hEAG1 is overexpressed in human osteosarcoma and the hEAG1 polyclonal antibody offers a good tool for further characterization of the oncogenic function of hEAG1 in osteosarcoma.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Ether-A-Go-Go Potassium Channels/genetics , Osteosarcoma/genetics , Recombinant Proteins/genetics , Adolescent , Adult , Aged , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Bone Neoplasms/metabolism , Child , Ether-A-Go-Go Potassium Channels/immunology , Ether-A-Go-Go Potassium Channels/metabolism , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoenzyme Techniques , Ion Channel Gating/drug effects , Male , Middle Aged , Osteosarcoma/metabolism , Prognosis , RNA, Messenger/genetics , Rabbits , Real-Time Polymerase Chain Reaction , Recombinant Proteins/metabolism , Tumor Cells, Cultured , Young Adult
9.
Immunobiology ; 217(1): 28-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22015046

ABSTRACT

Toxoplasma gondii is an intracellular parasite that has to cope with the microbicidal actions of IFNγ. Previously we reported that parasite-mediated induction of suppressor of cytokine signaling protein 1 (SOCS1) contributes to inhibition of IFNγ signaling. However, the signaling requirements remained elusive. We now show that induction of SOCS1 and inhibition of nitric oxide production by IFNγ was independent of stimulation of Toll-like receptors. Instead, infection by T. gondii resulted in induction of egr transcription factors which have been reported to regulate SOCS expression. Indeed, induction of egr2 as well as SOCS1 was dependent on p38 MAP kinase and blockade of egr inhibited SOCS1 expression. Moreover, we found that Mic8, a previously identified invasion factor of T. gondii, was necessary for SOCS1 regulation and escape of IFNγ mediated nitric oxide secretion within macrophages. Surprisingly, when further analyzing Mic8 deficient parasites we noted that inhibition of IFNγ mediated up-regulation of MHC-class II and ICAM1 molecules was independent of cell invasion. Furthermore, these inhibitory effects were equally observed in type I and II strains of T. gondii and were dependent on excreted and secreted antigens. In contrast, only the virulent RH type I strain additionally induced SOCS1 and efficiently inhibited nitric oxide secretion by IFNγ. The results show that T. gondii makes use of two different mechanisms to escape from IFNγ activity with one mode being strain dependent and relying on active cell invasion and SOCS1 induction.


Subject(s)
Gene Expression/immunology , Interferon-gamma/immunology , Macrophages/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Line , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/immunology , Genes, MHC Class II/immunology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Interferon-gamma/biosynthesis , Macrophages/metabolism , Macrophages/parasitology , Mice , Nitric Oxide/immunology , Nitric Oxide/metabolism , Protozoan Proteins/genetics , Species Specificity , Suppressor of Cytokine Signaling Proteins/genetics , Toxoplasma/metabolism , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Animal/parasitology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
10.
J Neurosci Res ; 88(8): 1651-63, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20077429

ABSTRACT

Campylobacteriosis is a frequent antecedent event in Guillain-Barré syndrome (GBS), inducing high-titer serum antibodies for ganglioside antigens in the peripheral nervous system (PNS). Molecular mimicry between the lipooligosaccharide (LOS) component of Campylobacter jejuni and human peripheral nerve gangliosides is believed to play an important role in the pathogenesis of GBS. Conventional treatment strategies for patients with GBS include plasmapheresis, intravenous immunoglobulin (IVIG), and immunosuppression, which are invasive or relatively ineffective. In this study, we used our animal model of GBS, in which Lewis rats were immunized with GD3-like LOS isolated from C.jejuni. The animals developed anti-GD3 ganglioside antibodies and manifested neuromuscular dysfunction. To develop novel therapeutic strategies, we treated the animals by intraperitoneal administration of an anti-GD3 antiidiotype monoclonal antibody (BEC2) that specifically interacts with the pathogenic antibody. The treated animals had a remarkable reduction of anti-GD3 antibody titers and improvement of motor nerve functions. The results suggest that ganglioside mimics, such as antiidiotype antibodies, may be powerful reagents for therapeutic intervention in GBS by neutralizing specific pathogenic antiganglioside antibodies.


Subject(s)
Antibodies, Anti-Idiotypic/therapeutic use , Gangliosides/immunology , Neuritis, Autoimmune, Experimental/therapy , Action Potentials/drug effects , Action Potentials/physiology , Animals , Biotinylation/methods , Campylobacter jejuni/immunology , Coculture Techniques/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Ether-A-Go-Go Potassium Channels/immunology , Ether-A-Go-Go Potassium Channels/metabolism , Ether-A-Go-Go Potassium Channels/pharmacokinetics , Female , Freund's Adjuvant/immunology , Lipopolysaccharides , Motor Neurons/pathology , Motor Neurons/ultrastructure , Muscle, Skeletal/physiology , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacokinetics , Neuritis, Autoimmune, Experimental/chemically induced , Neuritis, Autoimmune, Experimental/complications , Neuritis, Autoimmune, Experimental/immunology , Neuromuscular Junction Diseases/drug therapy , Neuromuscular Junction Diseases/etiology , Neuromuscular Junction Diseases/metabolism , Organ Culture Techniques , Rats , Rotarod Performance Test/methods , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure , Spinal Cord/physiology , Time Factors
11.
Cancer Res ; 68(6): 1611-3, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18339837

ABSTRACT

Emerging evidence indicates that ion channels act in a variety of physiologic and pathologic processes beyond electronic signal transmission, including in cancer. We recently found that the potassium channel Eag1 can mediate cancer progression and that a monoclonal antibody, which inhibits Eag1 action, can effectively restrict cancer cell proliferation. We discuss how Eag1 targeting may be useful in diagnostic or therapeutic settings.


Subject(s)
Antibodies, Monoclonal/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Neoplasms/therapy , Antibodies, Monoclonal/immunology , Drug Delivery Systems/methods , Ether-A-Go-Go Potassium Channels/immunology , Humans
12.
Pflugers Arch ; 456(1): 247-54, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18286302

ABSTRACT

The inhibition of the human ether-à-go-go-related (hERG) K+ channels is the major cause of long QT syndromes inducing fatal cardiac arrhythmias. Ergtoxin 1 (ErgTx1) belongs to scorpion-toxins, which are K+ channel-blockers, and binds to hERG channel with 1:1 stoichiometry and high affinity (Kd approximately 10 nM). Nevertheless, patch-clamp recordings recently demonstrated that ErgTx1 does not establish complete blockade of hERG currents, even at high ErgTx1 concentrations. Such phenomenon is supposed to be consistent with highly dynamic conformational changes of the outer pore domain of hERG. In this study, simultaneous topography and recognition imaging (TREC) on hERG HEK 293 cells was used to visualize binding sites on the extracellular part of hERG channel (on S1-S2 region) for Anti-Kv11.1 (hERG-extracellular-antibody). The recognition maps of hERG channels contained recognition spots, haphazardly distributed and organized in clusters. Recognition images after the addition of ErgTx1 at high concentrations ( approximately 1 microM) revealed subsequent partial disappearance of clusters, indicating that ErgTx1 was bound to the S1-S2 region. These results were supported by AFM force spectroscopy data, showing for the first time that voltage sensing domain (S1-S4) of hERG K+ channel might be one of the multiple binding sites of ErgTx1.


Subject(s)
Ether-A-Go-Go Potassium Channels/ultrastructure , Microscopy, Atomic Force/methods , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/ultrastructure , Scorpion Venoms/metabolism , Antibodies/immunology , Cell Line , Ether-A-Go-Go Potassium Channels/immunology , Humans , Kidney/cytology , Kidney/embryology , Kidney/ultrastructure , Protein Binding , Protein Structure, Tertiary
14.
Cancer Res ; 67(15): 7343-9, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671204

ABSTRACT

The potassium channel ether à go-go has been directly linked to cellular proliferation and transformation, although its physiologic role(s) are as of yet unknown. The specific blockade of human Eag1 (hEag1) may not only allow the dissection of the role of the channel in distinct physiologic processes, but because of the implication of hEag1 in tumor biology, it may also offer an opportunity for the treatment of cancer. However, members of the potassium channel superfamily are structurally very similar to one another, and it has been notoriously difficult to obtain specific blockers for any given channel. Here, we describe and validate the first rational design of a monoclonal antibody that selectively inhibits a potassium current in intact cells. Specifically blocking hEag1 function using this antibody inhibits tumor cell growth both in vitro and in vivo. Our data provide a proof of concept that enables the generation of functional antagonistic monoclonal antibodies against ion channels with therapeutic potential. The particular antibody described here, as well as the technique developed to make additional functional antibodies to Eag1, makes it possible to evaluate the potential of the channel as a target for cancer therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Ether-A-Go-Go Potassium Channels/immunology , Mammary Neoplasms, Experimental/therapy , Pancreatic Neoplasms/therapy , Potassium Channel Blockers/therapeutic use , Animals , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay , Electrophysiology , Enzyme-Linked Immunosorbent Assay , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Female , Fluorescent Antibody Technique , Humans , Kidney/cytology , Kidney/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, SCID , Mutagenesis, Site-Directed , Neuroblastoma/immunology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Transplantation, Heterologous
15.
Mol Cancer ; 5: 41, 2006 Oct 05.
Article in English | MEDLINE | ID: mdl-17022810

ABSTRACT

BACKGROUND: Certain types of potassium channels (known as Eag1, KCNH1, Kv10.1) are associated with the production of tumours in patients and in animals. We have now studied the expression pattern of the Eag1 channel in a large range of normal and tumour tissues from different collections utilising molecular biological and immunohistochemical techniques. RESULTS: The use of reverse transcription real-time PCR and specifically generated monoclonal anti-Eag1 antibodies showed that expression of the channel is normally limited to specific areas of the brain and to restricted cell populations throughout the body. Tumour samples, however, showed a significant overexpression of the channel with high frequency (up to 80% depending on the tissue source) regardless of the detection method (staining with either one of the antibodies, or detection of Eag1 RNA). CONCLUSION: Inhibition of Eag1 expression in tumour cell lines reduced cell proliferation. Eag1 may therefore represent a promising target for the tailored treatment of human tumours. Furthermore, as normal cells expressing Eag1 are either protected by the blood-brain barrier or represent the terminal stage of normal differentiation, Eag1 based therapies could produce only minor side effects.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Neoplasms/metabolism , Animals , Antibodies, Monoclonal , Antibody Specificity , Breast Neoplasms/metabolism , CHO Cells , Colonic Neoplasms/metabolism , Cricetinae , Cricetulus , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/immunology , Female , Humans , Immunohistochemistry , Liver Neoplasms/metabolism , Male , Neoplasms/genetics , Neoplasms/immunology , Prostatic Neoplasms/metabolism , RNA, Messenger/analysis , Respiratory Tract Neoplasms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Transfection , Up-Regulation
16.
FEBS Lett ; 580(12): 2850-2, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16783874

ABSTRACT

An increasing number of ion channels are being found to be causally involved in diseases, giving rise to the new field of "channelopathies". Cancer is no exception, and several ion channels have been linked to tumour progression. Among them is the potassium channel EAG (Ether-a-go-go). Over 75% of tumours have been tested positive using a monoclonal antibody specific for EAG, while inhibition of this channel decreased the proliferation of EAG expressing cells. The inhibition of EAG is accomplished using RNA interference, functional anti-EAG1 antibodies, or (unspecific) EAG channel blockers. Fluorescently labelled recombinant Fab fragments recognizing EAG allow the distribution of EAG to be visualized in an in vivo mouse tumour model.


Subject(s)
Biomarkers, Tumor/physiology , Ether-A-Go-Go Potassium Channels/physiology , Antibodies/immunology , Cell Cycle/physiology , Cell Proliferation , Ether-A-Go-Go Potassium Channels/immunology , Fluorescent Dyes , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...