KCNE1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
KCNE1; Potassium voltage-gated channel subfamily E member 1; Delayed rectifier potassium channel subunit IsK; IKs producing slow voltage-gated potassium channel subunit beta Mink; Minimal potassium channel
Target Names
KCNE1
Uniprot No.

Target Background

Function
KCNE1 is an ancillary protein that assembles as a beta subunit with a voltage-gated potassium channel complex, composed of pore-forming alpha subunits. It plays a crucial role in modulating the gating kinetics and enhancing the stability of the channel complex. When assembled with KCNB1, it modulates the gating characteristics of the delayed rectifier voltage-dependent potassium channel KCNB1. In combination with KCNQ1/KVLQT1, it is believed to form the slowly activating delayed rectifier cardiac potassium (IKs) channel, where the outward current reaches its steady state only after 50 seconds. Furthermore, when assembled with KCNH2/HERG, KCNE1 may modulate the rapidly activating component of the delayed rectifying potassium current in the heart (IKr).
Gene References Into Functions
  1. The S38G mutation in KCNE1 has been linked to a loss-of-function of IKs due to decreased KCNE1 protein expression and defects in KCNE1 protein membrane trafficking. PMID: 29395134
  2. Extensive molecular dynamics simulations have refined models of the interaction between the KCNQ1 channel and its accessory KCNE1 beta subunits. These analyses provide insights into the intricate relationship between these proteins. PMID: 28992529
  3. In lesion samples from children with cardiac insufficiency, significant differences in AA and CC genotype frequencies, as well as allele frequencies, were observed between the study group and the control group for the Mink-S27 variant. PMID: 28829496
  4. Sumoylation of KCNQ1 is dependent on KCNE1 and plays a critical role in determining the native characteristics of cardiac IKs in vivo. PMID: 28743749
  5. Equine KV7.1/KCNE1 expressed in CHO-K1 cells exhibits electrophysiological properties that are generally similar to their human counterparts. However, a slower deactivation was observed, potentially leading to a higher number of open channels at fast rates. PMID: 28917093
  6. The KCNE1 (rs1805127) variant appears to be an independent risk factor for atrial fibrillation (AF) in the Uygur population. Additionally, the KCNE4 (rs12621643) variant was identified as an independent risk factor for AF among both Uygur and Han individuals. PMID: 28981946
  7. Western blotting analysis, coupled with pharmacological data, suggests that long-term insulin treatment enhances KCNQ1/KCNE1 currents by increasing KCNE1 protein expression. PMID: 28882596
  8. While the KCNQ1/KCNE1 channel does not require phosphatidylinositol-4,5-bisphosphate (PIP2) or phosphatidylinositol-4-phosphate for anterograde trafficking, its function at the plasma membrane heavily relies on PIP2. PMID: 29020060
  9. Genetic variants rs426496 in AQP2, rs591810 in AQP3, and rs1805127, rs1805128, and rs17173510 in KCNE1 were identified in patients with Meniere's disease. PMID: 27509294
  10. Meta-analysis indicates that the G38S polymorphism in the KCNE1 gene significantly elevates the risk of atrial fibrillation in both Chinese and white individuals. PMID: 28640127
  11. Sphingomyelin synthase 1 positively regulates KCNQ1/KCNE1 channel density in a protein kinase D-dependent manner. PMID: 27194473
  12. Receptor Species-dependent Desensitization Controls KCNQ1/KCNE1 K+ Channels as Downstream Effectors of Gq Protein-coupled Receptors. PMID: 27834678
  13. KCNE1 encodes a modulator of both KCNQ1 and KCNH2 channels. Long QT syndrome (LQTS) patients carrying the KCNE1(G38S) variant exhibited a rate-dependent repolarization abnormality similar to patients with LQT2, indicating a potential risk for developing lethal arrhythmias. PMID: 27255646
  14. A stoichiometry of 4:4 has been established between the alpha KCNE1 and the beta KCNQ1 subunits. PMID: 26802629
  15. Based on current evidence from published studies, neither of the two variants from KCNE was significantly associated with the risk of Meniere's disease. PMID: 26890422
  16. Analysis of the QT-RR relationship can be utilized to evaluate the latent arrhythmogenicity of KCNE1(G38S). PMID: 26520166
  17. The incidence of AF among the senior Uygur population in Xinjiang territory was correlated with the KCNE1 (G38S) polymorphism, suggesting it may be an independent risk factor for Uygur AF patients. PMID: 26662381
  18. The KCNQ1 F279I mutation leads to a gain of function of IKs due to impaired gating modulation of Kv7.1 induced by KCNE1, resulting in a shortening of the cardiac action potential. PMID: 26168993
  19. This study aimed to assess the associations between polymorphisms in KCNE1, KCNQ1, and KCNH2 and the risk of AF in a Chinese population. PMID: 26066992
  20. KCNE1 and KCNE3 play distinct roles in the regulation of voltage-gated K(+) channels. PMID: 26410412
  21. Two phenylalanine residues on KCNQ1, Phe232 on S4 (VSD) and Phe279 on S5 (pore domain), have been identified as crucial for the gating modulation by KCNE1. PMID: 24920132
  22. Acute application of PUFAs increases Kv7.1/KCNE1, likely through a direct effect, and exhibits antiarrhythmic efficacy under IKr block. PMID: 25497550
  23. Phosphorylation of IKs and alpha1-AR regulation via activation of calcium-dependent PKC isoforms (cPKC) may be a key mechanism for controlling channel voltage-dependent activation and consequently action potential duration (APD) in response to adrenergic stimuli. PMID: 25479336
  24. The distal C-terminus of KCNE1 appears to be essential for the regulation of yotiao-mediated PKA phosphorylation. PMID: 25037568
  25. The KCNE1 112G polymorphism may be a risk factor for AF, and the 112G>A variant could potentially serve as a biomarker for predicting the development of AF. PMID: 25366730
  26. Klotho upregulates KCNQ1 and KCNE1 channel activity by enhancing channel protein abundance in the cell membrane. PMID: 24457979
  27. The KCNE1-D85N variant exhibits lower stability compared to the wild-type protein and undergoes rapid degradation through the ubiquitin-proteasome system. Verapamil may offer therapeutic value in LQTS patients by preventing the degradation of KCNE1-D85N. PMID: 24499369
  28. The resilience of the curvature within the KCNE1 transmembrane domain is likely to be maintained upon binding of the protein to the KCNQ1 channel. PMID: 25234231
  29. A significant association between the Mink S38G gene polymorphism and AF risk was observed. Carriers of the G allele may be predisposed to AF. PMID: 24696659
  30. The A590T mutation in the KCNQ1 C-terminal helix D diminishes KCNE1 channel trafficking and function but does not affect Yotiao interaction. PMID: 24713462
  31. Homozygous inheritance of KCNE1(38S) might lead to a mild reduction of the delayed rectifier K(+) currents, potentially increasing arrhythmogenic potential, especially in the presence of QT-prolonging factors. PMID: 24419801
  32. KCNE1 divides the voltage sensor movement in KCNQ1/KCNE1 channels into two distinct steps. PMID: 24769622
  33. The transmembrane domains (TMDs) of KCNE1 and KCNE2 have been shown to interact with the KCNQ1 channel in different modes. PMID: 24827085
  34. This study concluded that variants in the KCNQ1, KCNH2, KCNE1, and KCNE2 genes may be associated with the occurrence of certain cases of sudden unexplained nocturnal death syndrome in southern China. PMID: 23890619
  35. This study confirmed the association of the 112G>A polymorphism with postoperative AF in a cohort of patients undergoing cardiac surgery. PMID: 24439990
  36. Subjects with LQT-PM exhibited longer QTc intervals at rest and at peak exercise and during all phases of the recovery period compared to controls. Individuals with homozygous SNPs (KCNE1 35%) had longer resting QTc intervals compared to heterozygotes. PMID: 23714088
  37. The KCNE1 rs1805127 polymorphism increases the risk of atrial fibrillation. PMID: 23874724
  38. While KCNE1 maintained a stable cell surface localization, KCNQ1 exhibited variations in the cytosolic compartment (striations versus vesicles) and the degree of presence on the cell surface of cardiac myocytes. PMID: 24142691
  39. A single nucleotide polymorphism in KCNE1 was linked to tinnitus that developed independently of hearing loss. PMID: 23224734
  40. Abnormalities and mutations in the KCNE1 gene have been associated with the development of Long QT syndrome (LQTS). PMID: 23237912
  41. KCNE1 redirects the targeting-deficient KV7.1-Y51A mutant to the apical membrane of MDCK cells. PMID: 23324056
  42. Unsaturated heteromultimeric (KCNQ1)4(KCNE1)n channels exist as components of IKs and are pharmacologically distinct from KCNE-saturated KCNQ1-KCNE1 channels. PMID: 23650380
  43. KCNE1 and KCNE2, auxiliary subunits of voltage-gated potassium channels, undergo sequential cleavage mediated by either alpha-secretase and presenilin(PS)/gamma-secretase or BACE1 and PS/gamma-secretase in cells. PMID: 23504710
  44. Genetic polymorphisms in KCNE1 were associated with an increased risk of AF in a Chinese Han population. PMID: 23020083
  45. The KCNE1 G38S variant might have varying impacts on AF across different ethnicities. PMID: 23129484
  46. Genotype screening revealed the KCNE1 D85N polymorphism, which is recognized as one of the typical disease-causing gene variants in Long QT syndrome (LQTS). PMID: 22999324
  47. IKs channels open slowly because KCNE1 accessory subunits slow the movement of S4 voltage sensors in KCNQ1 pore-forming subunits. PMID: 23359697
  48. KCNQ1 channels, both in the absence and presence of KCNE1, undergo sequential gating transitions leading to channel opening even before all VSDs have moved. PMID: 22908235
  49. Early-onset lone atrial fibrillation has been linked to mutations in the potassium current channel regulatory subunit KCNE1. PMID: 22471742
  50. The KCNE1 variant rs1805128, resulting in D85N, confers a substantially increased risk for drug-induced torsades de pointes. PMID: 22100668

Show More

Hide All

Database Links

HGNC: 6240

OMIM: 176261

KEGG: hsa:3753

STRING: 9606.ENSP00000337255

UniGene: Hs.121495

Involvement In Disease
Jervell and Lange-Nielsen syndrome 2 (JLNS2); Long QT syndrome 5 (LQT5)
Protein Families
Potassium channel KCNE family
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Apical cell membrane. Membrane raft.
Tissue Specificity
Expressed in lung, kidney, testis, ovaries, small intestine, peripheral blood leukocytes. Expressed in the heart. Not detected in pancreas, spleen, prostate and colon. Restrictively localized in the apical membrane portion of epithelial cells.

Q&A

What is KCNE1 and why is it significant in research?

KCNE1 functions as a regulatory subunit of voltage-gated potassium (Kv) channel complexes, modulating gating kinetics and enhancing stability of channel complexes. It is particularly significant as it associates with KCNQ1/KVLQT1 alpha subunit to form the slowly activating delayed rectifier cardiac potassium (IKs) channel responsible for ventricular muscle action potential repolarization. This association makes KCNE1 critically important in cardiac physiology research and studies of certain cardiac arrhythmias. KCNE1 is also known by several alternative names including delayed rectifier potassium channel subunit IsK, minimal potassium channel (MinK), and IKs producing slow voltage-gated potassium channel subunit beta Mink .

What experimental applications are KCNE1 antibodies suitable for?

Commercially available KCNE1 antibodies, such as the rabbit polyclonal antibody ab65795, have been validated for multiple experimental techniques including Western Blot (WB), Immunohistochemistry on paraffin-embedded tissues (IHC-P), and Immunocytochemistry/Immunofluorescence (ICC/IF). These antibodies have been particularly validated for human samples, though cross-reactivity with other species may exist based on sequence homology. When selecting a KCNE1 antibody, researchers should verify the specific applications the antibody has been validated for and whether it has been cited in peer-reviewed publications .

How should proper controls be designed when using KCNE1 antibodies?

When designing experiments with KCNE1 antibodies, several controls are essential. Negative controls should include samples where KCNE1 is not expressed or has been knocked down. For immunoprecipitation experiments, controls should include untransfected cells, cells expressing only KCNQ1, cells expressing only KCNE1, and samples processed with control antibodies that shouldn't recognize KCNE1. For example, in co-immunoprecipitation studies examining KCNE1 C-terminus interactions with KCNQ1, researchers have used untransfected cells, KCNQ1 alone-expressing cells, KCNE1 wild-type alone-expressing cells, and KCNQ1+KCNE1 with control antibody samples as appropriate controls .

What are the optimal conditions for detecting KCNE1-KCNQ1 interactions?

The detection of KCNE1-KCNQ1 interactions requires careful experimental design. Co-immunoprecipitation experiments have been successfully conducted using antibodies against KCNQ1 to pull down the complex, followed by immunoblotting with anti-FLAG antibodies to detect tagged KCNE1 proteins. For functional studies examining KCNE1-KCNQ1 interactions, electrophysiological recordings in expression systems such as Xenopus oocytes have proven effective. When studying the time course of KCNE1-KCNQ1 association, measurements at multiple time points (2h, 4h, 6h, and 18h) post-protein introduction provide comprehensive data on the kinetics of interaction . Experiments should include appropriate controls and may benefit from complementary approaches such as FRET or proximity ligation assays.

What expression systems are most suitable for studying KCNE1 function with antibody-based techniques?

Multiple expression systems have been successfully employed for studying KCNE1 function. Xenopus oocytes offer advantages for electrophysiological studies combined with biochemical analysis, as demonstrated in experiments where purified recombinant KCNE1 was injected into oocytes expressing KCNQ1. Mammalian cell lines including HEK-293, CHO and COS-7 cells have also been effectively used to study KCNE1-KCNQ1 assembly and trafficking . When selecting an expression system, researchers should consider whether they need to study channel assembly, trafficking, or functional properties. For antibody-based detection, the choice of expression system may affect protein glycosylation patterns, which can influence antibody recognition. In Xenopus oocytes, studies have shown that recombinant KCNE1 protein undergoes different post-translational modifications compared to KCNE1 translated from cRNA within the same cells .

How can KCNE1 antibodies be used to investigate channel assembly and trafficking?

KCNE1 antibodies serve as powerful tools for investigating channel assembly and trafficking mechanisms. Research has demonstrated that KCNQ1-KCNE1 assembly occurs early in the secretory pathway, likely in the endoplasmic reticulum (ER). To study these processes, researchers can use a combination of techniques: (1) Immunofluorescence microscopy with organelle markers and KCNE1 antibodies to track localization; (2) Surface biotinylation combined with immunoprecipitation to measure plasma membrane expression; and (3) Brefeldin A (BFA) treatments to block anterograde trafficking and study the timing of assembly .

Studies have shown that BFA exposure prevents the appearance of IKs currents following prKCNE1 injection into Xenopus oocytes expressing KCNQ1, indicating that functional KCNQ1-KCNE1 channels require vesicular trafficking through the secretory pathway to reach the plasma membrane. This approach revealed that KCNQ1-KCNE1 channels are removed from the plasma membrane with a half-life of approximately 9 hours, similar to the ~11-hour turnover rate reported in COS-7 cells .

What are the key considerations when using KCNE1 antibodies to study mutant forms associated with Long QT Syndrome?

When studying KCNE1 mutants associated with Long QT Syndrome (LQT5), antibody selection and experimental design require special attention. The proximal portion of the C-terminal domain of KCNE1 contains at least 9 known LQT5 mutations, indicating this region's critical importance for channel function . Researchers should consider whether mutations might affect antibody epitope recognition, potentially leading to false-negative results.

For functional studies of mutations, combining antibody-based detection of expression and localization with electrophysiological recordings provides comprehensive data. Deletion mutants like Y94stop (lacking amino acids 94-129) and N79stop (lacking amino acids 79-129) have revealed that the C-terminal domain of KCNE1 is critical for cAMP-dependent functional regulation of KCNQ1 . When studying such mutants, researchers should verify proper membrane expression and assembly with KCNQ1 before attributing functional changes to the specific mutations rather than to trafficking defects.

How can researchers use KCNE1 antibodies to investigate post-translational modifications and their functional implications?

KCNE1 undergoes various post-translational modifications that can be studied using specific antibodies. Glycosylation patterns can be investigated using glycosidase treatments followed by Western blotting with KCNE1 antibodies. Studies in Xenopus oocytes have shown different banding patterns between KCNE1 expressed from cRNA versus recombinant protein, indicating differences in glycosylation .

For phosphorylation studies, researchers can employ phospho-specific antibodies in combination with treatments that alter cellular phosphorylation states. While KCNQ1 is phosphorylated at Ser27 in response to cAMP independent of co-expression with KCNE subunits, the functional response of the channel complex to this phosphorylation depends on the presence of KCNE1 . Experimental approaches can include PKA activators (cAMP, forskolin) and phosphatase inhibitors (okadaic acid) coupled with phospho-specific antibodies and functional assays to correlate biochemical changes with alterations in channel properties.

What are common pitfalls when using KCNE1 antibodies, and how can they be avoided?

Several common pitfalls can affect experiments using KCNE1 antibodies. First, KCNE1's relatively small size (~15 kDa) can make it challenging to detect on Western blots, especially when glycosylated forms create diffuse bands. Second, many commercial antibodies may cross-react with other KCNE family members due to sequence similarities. Third, low expression levels in native tissues can lead to weak signals and false negatives.

To avoid these issues, researchers should: (1) Use fresh samples with appropriate protease inhibitors to prevent degradation; (2) Validate antibody specificity using overexpression systems and knockout/knockdown controls; (3) Optimize protein extraction conditions, considering that KCNE1 is a membrane protein requiring appropriate detergents; and (4) Consider using epitope-tagged KCNE1 constructs in heterologous expression systems where native antibodies yield poor results, following thorough validation that the tag doesn't interfere with function or localization.

How should researchers interpret conflicting data from antibody-based versus functional assays of KCNE1?

When antibody-based detection and functional assays yield conflicting results, several potential explanations should be considered. First, functional assays might detect even small amounts of properly assembled channels that are below the detection threshold of antibody-based methods. Second, antibodies might recognize non-functional or improperly folded proteins. Third, post-translational modifications might affect antibody recognition without altering function, or vice versa.

Resolution strategies include: (1) Employing multiple antibodies targeting different epitopes; (2) Using complementary detection methods such as mass spectrometry; (3) Verifying antibody specificity with appropriate controls; and (4) Considering the sensitivity limitations of each technique. In the case of mutant studies, researchers should determine whether discrepancies result from altered trafficking, assembly, or intrinsic channel properties by employing both biochemical and functional approaches in parallel.

What is the optimal approach for quantifying KCNE1 expression levels in different cellular compartments?

Quantifying KCNE1 expression in different cellular compartments requires careful experimental design. For total protein quantification, Western blot with appropriate loading controls provides relative expression levels. For membrane versus intracellular distribution, surface biotinylation followed by pull-down of biotinylated proteins can isolate plasma membrane proteins, which can then be analyzed by Western blotting with KCNE1 antibodies.

For subcellular localization studies, confocal microscopy with KCNE1 antibodies co-stained with organelle markers (ER, Golgi, plasma membrane) provides spatial information. Quantitative analysis should include multiple cells from independent experiments, with appropriate controls for antibody specificity and background subtraction. When comparing wild-type and mutant KCNE1, parallel processing and imaging using identical acquisition parameters are essential for valid comparisons of localization patterns and expression levels.

How might KCNE1 antibodies facilitate investigation of dynamic channel assembly in live cells?

Future applications of KCNE1 antibodies could enable real-time visualization of channel assembly dynamics in living cells. This might be achieved through development of antibody fragments (Fab, scFv) conjugated to cell-permeable fluorescent probes that retain specificity for KCNE1 without disrupting function. Complementary approaches include knock-in of epitope tags recognized by cell-permeable fluorescent antibodies or development of conformation-specific antibodies that selectively recognize assembled channel complexes versus unassembled subunits.

Studies have already demonstrated that KCNE1 and KCNQ1 assembly can occur independent of protein translation and that functional channel complexes assemble early in the secretory pathway, with a half-time of approximately 3 hours for functional association . Building on these findings, real-time imaging approaches could reveal whether assembly occurs continuously or at specific cellular checkpoints, providing insights into regulatory mechanisms of channel complex formation.

What role might KCNE1 antibodies play in understanding the structural basis of channelopathies?

KCNE1 antibodies could significantly advance understanding of the structural basis of channelopathies by enabling purification of native channel complexes for structural studies. Conformation-specific antibodies might allow isolation of channels in different functional states for cryo-electron microscopy analysis. Additionally, epitope mapping with a panel of KCNE1 antibodies could help identify critical interaction surfaces between KCNE1 and alpha subunits.

Research has already identified that the C-terminal domain of KCNE1 contains numerous Long QT Syndrome mutations and is critical for cAMP-dependent functional regulation . Advanced structural studies facilitated by specific antibodies could reveal how these mutations disrupt protein-protein interactions or alter channel gating mechanisms, potentially leading to new therapeutic approaches targeting specific molecular defects in channelopathies.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.