KCNJ2 Antibody

Shipped with Ice Packs
In Stock

Description

Target Antigen and Biological Role

The KCNJ2 antibody specifically binds to the Kir2.1 channel, a key inward-rectifier potassium channel. Kir2.1 stabilizes resting membrane potentials and modulates action potential duration in excitable cells . Mutations in KCNJ2 are linked to arrhythmias such as:

  • Andersen-Tawil syndrome (ATS1/LQT7)

  • Catecholaminergic polymorphic ventricular tachycardia 3 (CPVT3)

  • Short QT syndrome (SQT3)

  • Familial atrial fibrillation (FAF) .

Mechanistic Insights into Arrhythmias

  • The R67Q mutation in KCNJ2 reduces Kir2.1 current density under beta-adrenergic stimulation, contributing to CPVT3 .

  • Val302del and E299V mutations impair channel function, leading to ATS1 and altered cardiac excitability .

Cancer Research

  • In small-cell lung cancer (SCLC), KCNJ2 regulates drug resistance via MRP1/ABCC1. Knockdown via KCNJ2 shRNA sensitizes cancer cells to chemotherapy .

Neurological Studies

  • Kir2.1 is expressed in mouse midbrain neurons, with surface detection validated using APC-159 in live THP-1 cells .

Validation and Experimental Data

  • APC-026 demonstrated specificity in canine myocytes, with staining eliminated by pre-incubation with antigenic peptide .

  • APC-159 confirmed Kir2.1 surface expression in live human monocytic cells, critical for studying channel trafficking .

Clinical Relevance

KCNJ2 antibodies enable:

  • Diagnostics: Identifying KCNJ2 mutations in CPVT3 and ATS1 patients .

  • Therapeutic Development: Screening drugs targeting Kir2.1 dysfunction in arrhythmias .

  • Mechanistic Studies: Probing PKA-dependent channel phosphorylation in adrenergic-stimulated arrhythmias .

Key Limitations

  • Tissue Specificity: Kir2.1 expression varies between cardiac chambers, requiring validation in target tissues .

  • Mutation Effects: Heterozygous mutations (e.g., R67Q) may require co-expression studies to assess dominant-negative effects .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery times may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributor.
Synonyms
KCNJ2; IRK1; Inward rectifier potassium channel 2; Cardiac inward rectifier potassium channel; Inward rectifier K(+ channel Kir2.1; IRK-1; hIRK1; Potassium channel, inwardly rectifying subfamily J member 2
Target Names
KCNJ2
Uniprot No.

Target Background

Function
KCNJ2, encoding the inward rectifier potassium channel Kir2.1, plays a crucial role in regulating the electrical activity of cells. This channel is responsible for establishing the resting membrane potential and action potential waveform, particularly in neuronal and muscle tissues. Kir2.1 exhibits a preference for allowing potassium ions to flow into the cell, rather than out of it, a property known as inward rectification. The voltage dependence of the channel is influenced by extracellular potassium concentration, where increasing external potassium shifts the voltage range for channel opening towards more positive values. The inward rectification mechanism is primarily attributed to the blockage of outward current by intracellular magnesium. Notably, Kir2.1 can be blocked by extracellular barium or cesium.
Gene References Into Functions
  1. Genotyping of 12 tag single nucleotide polymorphisms (SNPs) from the KCNJ2 and KCNJ10 genes revealed a significant association between KCNJ10 rs1186689 and disease susceptibility. The variant T allele was associated with a lower risk of developing Autism Spectrum Disorder (ASD). PMID: 30304693
  2. Experimental data indicate that hydrocinnamic acid can inhibit Kir2.1 channel currents in both excised inside-out and whole-cell patch clamp configurations, with IC50 values of 5.21 +/- 1.02 mM and 10.08 +/- 0.46 mM, respectively. PMID: 28660286
  3. A study confirmed the pathogenicity of the Kir2.1-52V mutation in a patient with long-QT syndrome. This study also supports the use of isogenic human induced pluripotent stem cell-derived cardiomyocytes as a relevant model for screening variants of unknown function. PMID: 29021306
  4. A novel KCNJ2 sequence variant (p.Y145C) was identified in a family diagnosed with Andersen-Tawil syndrome. PMID: 29017447
  5. Combined inhibition of IKr and IKur currents produced a synergistic anti-arrhythmic effect in both forms of SQT3 (short QT syndrome 3). These findings suggest potential pharmacological strategies for managing SQT3-linked atrial fibrillation. PMID: 28609477
  6. An R204A mutation in Kir2.1 disrupts the characteristic cytoplasmic domain subunit interface salt bridges, leading to a reduced apparent sensitivity of channel activity to the ligand phosphatidylinositol bisphosphate (PIP2). PMID: 28446610
  7. Research suggests a significant role for KCNJ2 in the pathophysiology of Thyrotoxic Periodic Paralysis (TPP) in Korean Graves' Disease patients with TPP. PMID: 28008586
  8. Binding of the Nav1.5 N-terminal domain to alpha1-syntrophin enhances the membrane density of human Kir2.1, Kir2.2, and Nav1.5 channels. PMID: 26786162
  9. Kir2.1 may participate in macrophage maturation and differentiation and play a key role in lipid uptake and foam cell formation through modulation of scavenger receptor expression. PMID: 26689595
  10. A Korean family with Andersen-Tawil syndrome exhibiting a G215D mutation in the KCNJ2 gene was identified through diagnostic exome sequencing. PMID: 26927354
  11. Chloroethylclonidine interacts with Kir2.1 channels in the cytoplasmic pore. PMID: 26922543
  12. Variability was found in a three-generation family with Pierre Robin sequence, acampomelic campomelic dysplasia, and intellectual disability due to a novel approximately 1 Mb deletion upstream of SOX9, encompassing KCNJ2 and KCNJ16. PMID: 26663529
  13. Patients with Dilated Cardiomyopathy and Sustained Monomorphic Ventricular Tachycardia demonstrate up-regulation of the KCNN3 and KCNJ2 genes and CACNG8-linked left ventricular dysfunction. PMID: 26710323
  14. Silencing BKCa (KCa1.1) inhibits cell mobility, while silencing IKir (Kir2.1) increases cell mobility in human cardiac c-kit+ progenitor cells. PMID: 26390131
  15. No genetic variants were identified in the KCNJ2 gene in a cohort of Chinese thyrotoxic periodic paralysis patients. PMID: 25885757
  16. Kir2.1 channels exhibit a binding site determined by Cys311, responsible for drug-induced increases in inward rectifier Kir2.1 currents. PMID: 25205296
  17. Kir2.1 channel function is essential during osteoblastogenesis. PMID: 25205110
  18. Upregulation of the inwardly rectifying potassium channel Kir2.1 (KCNJ2) modulates multidrug resistance in small-cell lung cancer under the regulation of miR-7 and the Ras/MAPK pathway. PMID: 25880778
  19. Research suggests that Kir2.1 currents control the interspike interval, and blocking Kir2.1 channels increases the action potential frequency, which in turn enhances the rate of insulin secretion in pancreatic beta cells. PMID: 25727015
  20. The K346T mutation causes a gain of function in Kir2.1 channels by altering their trafficking and stabilization, suggesting a combined effect on cardiac rhythm and neuropsychiatric phenotype. PMID: 24794859
  21. This study demonstrated that KCNJ2 mutations cause a variable phenotype, with dysmorphic features observed in all patients studied, a high penetrance of periodic paralysis in males, and ventricular arrhythmia with a risk of sudden cardiac death. PMID: 24861851
  22. The R67Q-Kir2.1 mutation is associated with an adrenergic-dependent clinical and cellular phenotype, with rectification abnormality enhanced by increased calcium. PMID: 24561538
  23. Genetic variation in the KCNJ2 gene is identified as a significant locus for thyrotoxic periodic paralysis. PMID: 23803013
  24. Mutations in KCNJ2 have been linked to familial atrial fibrillation. PMID: 24460807
  25. SGK3 is identified as a novel regulator of K(ir)2.1. PMID: 24556932
  26. Multiple proteolytic pathways control Kir2.1 levels at the plasma membrane. PMID: 24227888
  27. This review describes the loss-of-function mutations in KCNJ2 associated with type 1 Andersen-Tawil syndrome. PMID: 24383070
  28. Despite a severe clinical presentation with a high rate of ventricular arrhythmias, the arrhythmic prognosis of Andersen-Tawil syndrome patients with KCNJ2 mutations is relatively favorable under treatment. PMID: 23867365
  29. Andersen-Tawil syndrome (ATS) is a rare inherited multisystem disorder associated with mutations in KCNJ2. PMID: 24047492
  30. This review compiles clinical, genetic, biochemical, electrophysiological, and molecular evidence identifying Kir2.1 as a molecular target for Fetal Alcohol Spectrum Disorder (FASD) development and potentially therapeutic treatment. PMID: 23756044
  31. Genetic background of catecholaminergic polymorphic ventricular tachycardia in Japan. PMID: 23595086
  32. A pentamidine analog is reported to specifically block the cardiac KIR2.1 channel, leading to a prolonged action potential duration. PMID: 23625347
  33. Kir2.1 gene expression inhibits motor neuron activity by resisting depolarization to the action potential threshold. PMID: 23277370
  34. Energetics and location of phosphoinositide binding in human Kir2.1 channels. PMID: 23564459
  35. Overexpression of the Kir2.1 channel attenuates the post-transplantation pro-arrhythmic risk in myocardial infarction. PMID: 23041574
  36. Kir2.1 loss of function is additive to the increase in late sodium current, prolonging repolarization and leading to arrhythmia generation in Cav3-mediated long QT syndrome 9. PMID: 23640888
  37. Two novel, heterozygous KCNJ2 mutations (p.N318S, p.W322C) located in the C-terminus of the Kir2.1 subunit were identified in Andersen-Tawil syndrome. PMID: 23644778
  38. Protein kinase B (PKB) in conjunction with PIKfyve activates Kir2.1 channels. PMID: 23188060
  39. miR-26 controls the expression of KCNJ2 and may play a role in atrial fibrillation. PMID: 23543060
  40. KCNJ2 mutations in short QT syndrome 3 result in atrial fibrillation and ventricular pro-arrhythmia. PMID: 23440193
  41. Mutations of two distant Kir2.1 cytosolic residues, Leu-222 and Asn-251, form a two-way molecular switch that controls channel modulation by cholesterol. PMID: 22995912
  42. The targeting of the K(ir)2.1 3'UTR at bp 2677-2683 by miR-212 results in a decrease in the red/green fluorescence intensity ratio, as determined by automated image analysis, confirming that mutations at this site alter the binding. PMID: 22880819
  43. The common variant rs7219669 is associated with the TPE interval (EKG) and is a candidate to modify repolarization-related arrhythmia susceptibility. PMID: 22342860
  44. Expression quantitative trait locus (eQTL) analysis identified SNPs in the region flanking rs312691 (+/-10 kb) that could potentially affect KCNJ2 expression in thyrotoxic periodic paralysis. PMID: 22863731
  45. Mutation-positive rates of KCNJ2 were 75% in typical ATS, 71% in cardiac phenotype alone, 100% in periodic paralysis, and 7% in CPVT (catecholaminergic polymorphic ventricular tachycardia). PMID: 22589293
  46. The KCNJ2-D172N mutation leads to accelerated ventricular repolarization and QT interval shortening, facilitating the initiation and maintenance of re-entrant circuits. PMID: 22308236
  47. Characterization of a novel, dominant negative KCNJ2 mutation associated with Andersen-Tawil syndrome. PMID: 22186697
  48. The C-terminal region is a vital motif for phosphatidylinositol-4,5-bisphosphate (PIP2) binding and channel trafficking, and defects in PIP2 binding have been reported to constitute a pathogenic mechanism in ATS. PMID: 21875779
  49. Bacterial Kir channels (KirBac1.1 and KirBac3.1) and human Kir2.1 are all inhibited by cholesterol, most likely by locking the channels into prolonged closed states, whereas the enantiomer, ent-cholesterol, does not inhibit these channels. PMID: 21559361
  50. A tetrad complex demonstrates the close association of the Kir2.1 cytoplasmic domains and the influence of PSD-95 mediated self-assembly on the clustering of these channels. PMID: 21756874

Show More

Hide All

Database Links

HGNC: 6263

OMIM: 170390

KEGG: hsa:3759

STRING: 9606.ENSP00000243457

UniGene: Hs.1547

Involvement In Disease
Long QT syndrome 7 (LQT7); Short QT syndrome 3 (SQT3); Atrial fibrillation, familial, 9 (ATFB9)
Protein Families
Inward rectifier-type potassium channel (TC 1.A.2.1) family, KCNJ2 subfamily
Subcellular Location
Membrane; Multi-pass membrane protein. Membrane; Lipid-anchor.
Tissue Specificity
Heart, brain, placenta, lung, skeletal muscle, and kidney. Diffusely distributed throughout the brain.

Q&A

What is KCNJ2 and why is it important in research?

KCNJ2 encodes the Kir2.1 protein, an inwardly rectifying potassium channel that allows potassium ions to move more easily into rather than out of cells. This channel is critically important in establishing highly negative resting membrane potentials and contributes to the long-lasting action potential plateau in various cells, particularly cardiac myocytes . Kir2.1 is expressed predominantly in skeletal and cardiac muscle tissues, making it a significant target for cardiovascular research . The inward rectification property of Kir2.1 is governed by intracellular ions such as Mg²⁺ and polyamines . Mutations in KCNJ2 are associated with Andersen syndrome (characterized by cardiac arrhythmias, periodic paralysis, and dysmorphic features) and Catecholaminergic Polymorphic Ventricular Tachycardia type 3 (CPVT3) . These disease associations make KCNJ2 antibodies valuable tools for both basic science and translational research.

What types of KCNJ2 antibodies are available for research?

Researchers have access to several types of KCNJ2/Kir2.1 antibodies that target different epitopes of the protein:

  • Extracellular domain antibodies: These antibodies target epitopes in the extracellular loop of Kir2.1. For example, antibodies targeting amino acid residues 112-125 of rat Kir2.1 are available and can be used for live cell applications .

  • C-terminal antibodies: Antibodies targeting the C-terminal region, such as those recognizing amino acids 401-427 of human KCNJ2, are useful for various applications including Western blotting and immunohistochemistry .

  • Different host species: KCNJ2 antibodies are produced in various host animals including rabbit (polyclonal) and mouse (monoclonal) .

  • Conjugated antibodies: Available conjugates include HRP, biotin, PE, FITC, and PerCP for specialized applications like flow cytometry .

The choice of antibody depends on the specific experimental requirements, including the target species, application method, and whether live cell studies are needed.

How can I validate the specificity of KCNJ2 antibodies?

Validating antibody specificity is critical for obtaining reliable experimental results. Recommended validation approaches include:

  • Blocking peptide experiments: Pre-incubate the antibody with its immunizing peptide before application. This should eliminate or significantly reduce specific staining, as demonstrated in mouse midbrain sections where pre-incubation of Anti-Kir2.1 antibody with its blocking peptide suppressed staining .

  • Western blot analysis: Analyze multiple tissue samples with known differential expression of KCNJ2. For example, comparing rat brain, rat heart, and mouse heart lysates can help confirm specificity across species and tissues .

  • Positive and negative controls: Include tissues or cell lines known to express high levels of Kir2.1 (such as cardiac tissue) as positive controls, and those with minimal expression as negative controls.

  • Knockout/knockdown validation: When available, use KCNJ2 knockout or knockdown samples to confirm antibody specificity.

  • Multiple antibody approach: Use antibodies targeting different epitopes of KCNJ2 and compare the results to improve confidence in specificity.

What are the optimal sample preparation methods for KCNJ2 antibody applications?

Sample preparation varies by application:

  • For Western blot:

    • Proper tissue or cell lysis using buffers containing protease inhibitors

    • Use of reducing conditions

    • Appropriate protein loading (typically 20-50 μg of total protein)

    • Recommended dilution for many KCNJ2 antibodies is 1:200

  • For immunohistochemistry:

    • Perfusion-fixed frozen sections yield excellent results for brain tissue

    • For paraffin-embedded sections, antigen retrieval may be necessary

    • Blocking of non-specific binding sites is critical

  • For live cell applications:

    • Use antibodies targeting extracellular epitopes

    • Apply primary antibody to intact cells (typically at 1:50 dilution)

    • Follow with fluorophore-conjugated secondary antibody

    • Maintain physiological conditions during the procedure

  • For flow cytometry:

    • Single-cell suspensions with minimal cellular debris

    • Appropriate blocking to prevent non-specific binding

    • Use of viability dyes to exclude dead cells from analysis

How can KCNJ2 antibodies be used to investigate cardiac channelopathies?

KCNJ2 mutations have been associated with several cardiac channelopathies, including Andersen syndrome and CPVT3. KCNJ2 antibodies can be valuable tools in investigating these conditions:

  • Mutation-specific protein expression analysis:

    • Compare Kir2.1 protein expression and localization in cells expressing wild-type versus mutant KCNJ2

    • Identify altered trafficking patterns in mutations such as R82W, V227F, R67W, C101R, G144D, T305S, and R260P that have been linked to CPVT3

    • Correlate protein expression with functional changes

  • Co-immunoprecipitation studies:

    • Use KCNJ2 antibodies to identify altered protein-protein interactions in disease states

    • Investigate potential interactions with calcium handling proteins like RYR2 and CASQ2, which are also implicated in CPVT

  • Cell surface expression quantification:

    • Use extracellular epitope antibodies to quantify changes in surface expression of wild-type versus mutant Kir2.1 channels

    • Employ surface biotinylation in combination with KCNJ2 antibodies to measure trafficking defects

  • Patient-derived cell studies:

    • Apply KCNJ2 antibodies to analyze channel expression in iPSC-derived cardiomyocytes from patients with KCNJ2 mutations

    • Compare localization patterns between patient and control samples

What methodologies are optimal for studying KCNJ2 in human iPSC-derived cardiomyocytes?

Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) represent a valuable model for studying cardiac ion channels, including KCNJ2. Optimal methodologies include:

  • KCNJ2 overexpression systems:

    • Overexpression of KCNJ2 in iPSC-CMs enhances their maturation, yielding increased Kir2.1 protein expression and current density

    • This approach produces more physiologically relevant cardiomyocytes with hyperpolarized maximal diastolic potential, shortened action potential duration, and increased maximal upstroke velocity

  • Immunofluorescence techniques:

    • Use KCNJ2 antibodies to track changes in channel expression during cardiomyocyte maturation

    • Combine with markers of cardiomyocyte structure (e.g., sarcomeric proteins) to correlate Kir2.1 expression with structural maturity

    • KCNJ2 overexpression leads to enlarged cell size and more elongated cell shape, indicating structural maturation

  • Correlation of protein expression with electrophysiology:

    • Combine patch-clamp recordings with immunostaining to correlate IK1 current density with Kir2.1 protein levels

    • Use barium chloride (BaCl₂), an IK1 blocker, to confirm the functional significance of observed currents

  • Engineered heart tissue (EHT) applications:

    • KCNJ2 overexpressing iPSC-CMs form more mature human EHTs with improved tissue structure and cell junctions

    • Apply KCNJ2 antibodies to analyze channel distribution within these 3D tissue constructs

How can KCNJ2 antibodies be integrated with electrophysiological studies?

Integrating immunodetection with functional electrophysiology provides powerful insights into ion channel function:

  • Patch-clamp with immunocytochemistry:

    • Perform patch-clamp recordings followed by immunostaining of the same cell

    • Mark recorded cells for subsequent immunodetection using fluorescent dyes or grid-marked coverslips

    • Correlate current amplitude with protein expression level

  • Subcellular localization studies:

    • Use high-resolution confocal microscopy with KCNJ2 antibodies to map channel distribution

    • Correlate with local electrophysiological properties using techniques like scanning ion conductance microscopy

  • Heterologous expression systems:

    • Transfect cells with wild-type or mutant KCNJ2 constructs

    • Quantify surface expression using extracellular epitope antibodies

    • Directly correlate with whole-cell patch-clamp measurements of IK1

  • Pharmacological manipulation:

    • Apply channel modulators like barium chloride to block IK1

    • Compare electrophysiological effects with changes in antibody accessibility or binding

What are the best approaches for multiplexed detection of KCNJ2 with other cardiac ion channels?

Cardiac function depends on the coordinated action of multiple ion channels. Approaches for multiplexed detection include:

  • Multi-color immunofluorescence:

    • Use spectrally distinct fluorophores conjugated to antibodies against different ion channels

    • Select compatible antibodies raised in different host species to avoid cross-reactivity

    • Kir2.1 can be co-detected with calcium handling proteins (RYR2, CASQ2) implicated in related channelopathies

  • Sequential immunostaining protocols:

    • Apply and detect the first antibody, then strip or quench the signal

    • Apply subsequent antibodies in sequence

    • Document each step with appropriate imaging

  • Proximity ligation assays:

    • Detect protein-protein interactions between KCNJ2 and other channel proteins

    • Generate fluorescent signals only when target proteins are within 30-40 nm of each other

    • Useful for identifying channel complexes and regulatory interactions

  • Flow cytometry:

    • Use antibodies against extracellular epitopes of multiple channels

    • Apply to dissociated cells for quantitative analysis of co-expression patterns

    • Particularly valuable for sorting cell populations based on channel expression profiles

How to design experiments that distinguish between KCNJ2 expression changes versus functional alterations?

Distinguishing between changes in channel expression and functional alterations requires carefully designed experiments:

  • Combined protein quantification and functional assessment:

    • Quantify total and surface protein expression using Western blot and surface biotinylation

    • Compare with functional measurements using patch-clamp or potassium flux assays

    • Discrepancies between expression and function may indicate post-translational modifications or altered channel properties

  • Single-cell correlation studies:

    • Perform functional recordings on individually identified cells

    • Follow with immunocytochemistry on the same cells

    • Analyze correlation between function and expression at the single-cell level

  • Pharmacological distinction:

    • Apply specific IK1 blockers like barium chloride at different concentrations

    • Compare inhibition profiles between experimental conditions

    • Altered sensitivity may indicate changes in channel properties rather than expression levels

  • Mutational analysis:

    • Introduce specific mutations that alter channel function but not trafficking

    • Use antibodies to confirm equivalent surface expression

    • Attribute functional differences to altered channel properties

What are the common pitfalls when using KCNJ2 antibodies and how to avoid them?

Several technical issues can compromise experiments with KCNJ2 antibodies:

  • Non-specific binding:

    • Always include appropriate negative controls

    • Use blocking peptides to confirm specificity

    • Test antibodies on samples known to lack KCNJ2 expression

    • Optimize antibody concentration (typically 1:200 for Western blot, 1:50 for immunocytochemistry)

  • Cross-reactivity with other Kir channels:

    • Kir channel family members share sequence homology

    • Validate antibodies against cells expressing single channel subtypes

    • Consider using multiple antibodies targeting different epitopes

  • Membrane protein solubilization issues:

    • Use appropriate detergents for membrane protein extraction

    • Avoid excessive heating which may cause protein aggregation

    • Consider non-reducing conditions if antibody recognition depends on disulfide bonds

  • Fixation-dependent epitope masking:

    • Test multiple fixation protocols (paraformaldehyde, methanol, acetone)

    • Optimize antigen retrieval methods for fixed tissues

    • For some epitopes, consider using live cell staining with extracellular antibodies

How to optimize KCNJ2 antibody-based assays for different species?

KCNJ2 is conserved across species but with sequence variations that affect antibody binding:

  • Species-specific epitope selection:

    • Check sequence homology of the antibody epitope across target species

    • For cross-species studies, select antibodies targeting highly conserved regions

    • Available KCNJ2 antibodies have been validated for human, rat, and mouse samples

  • Validation across species:

    • Test antibodies on positive control samples from each species of interest

    • For Western blot, compare band patterns between species (e.g., rat brain, rat heart, and mouse heart)

    • Adjust antibody concentration for each species

  • Secondary antibody considerations:

    • Select secondary antibodies specific to the host species of the primary antibody

    • Ensure secondary antibodies have minimal cross-reactivity with proteins from the target species

    • For multi-species studies, consider directly conjugated primary antibodies

  • Predicted reactivity:

    • Some commercially available antibodies have predicted reactivity with additional species (e.g., bovine, canine, pig, rabbit, rat)

    • Always validate experimentally before conducting critical experiments

What approaches are recommended for quantifying KCNJ2 expression levels?

Accurate quantification of KCNJ2 expression requires appropriate methods:

  • Western blot quantification:

    • Include loading controls (housekeeping proteins or total protein stains)

    • Generate standard curves using recombinant proteins if absolute quantification is needed

    • Use digital imaging and analysis software for densitometry

    • Normalize to appropriate references depending on experimental question

  • Flow cytometry:

    • Use antibodies targeting extracellular epitopes for intact cell analysis

    • Include calibration beads to standardize fluorescence intensity

    • Calculate molecules of equivalent soluble fluorochrome (MESF) for cross-experiment comparison

    • Analyze median fluorescence intensity rather than mean for non-normal distributions

  • Quantitative immunofluorescence:

    • Include internal standards in each experiment

    • Use identical acquisition parameters for all samples

    • Apply background subtraction and flat-field correction

    • Consider automated image analysis to reduce bias

  • qPCR correlation:

    • Combine protein quantification with mRNA measurement

    • Assess correlation between transcript and protein levels

    • Identify potential post-transcriptional regulatory mechanisms

How are KCNJ2 antibodies being used in drug discovery and cardiotoxicity testing?

KCNJ2 antibodies facilitate drug development and safety assessment:

  • High-throughput screening platforms:

    • Antibody-based assays to measure changes in channel expression or localization in response to compounds

    • Correlation with functional effects in automated patch-clamp systems

    • KCNJ2 overexpressing iPSC-CMs provide a more stable platform for drug testing, allowing for detection of significant drug responses in concentration-dependent manner

  • Cardiotoxicity prediction:

    • Monitor changes in Kir2.1 expression in response to compounds

    • Correlate with electrophysiological parameters like action potential duration

    • For example, testing with hERG blocker E-4031 in KCNJ2 overexpressing iPSC-CMs showed concentration-dependent APD prolongation

  • Engineered tissue models:

    • Apply KCNJ2 antibodies to analyze channel distribution in 3D cardiac tissues

    • Assess drug effects on channel expression and localization

    • KCNJ2 overexpression improves tissue structure and cell junctions in engineered heart tissues

  • Patient-specific drug response prediction:

    • Use antibodies to characterize channel expression in patient-derived iPSC-CMs

    • Correlate with drug sensitivity to develop personalized treatment approaches

    • Particularly valuable for patients with KCNJ2 mutations

What is the role of KCNJ2 in cardiac development and maturation?

KCNJ2 plays a crucial role in cardiac development and maturation:

  • Developmental expression patterns:

    • KCNJ2 expression increases during cardiomyocyte maturation

    • Expression in iPSC-CMs is significantly lower than in adult human left ventricular tissues

    • Antibodies can track this developmental progression

  • Functional consequences of KCNJ2 expression:

    • KCNJ2 overexpression in iPSC-CMs enhances electrophysiological maturation

    • Results in hyperpolarized maximal diastolic potential, shortened action potential duration, and increased maximal upstroke velocity

    • Leads to more mature Ca²⁺ signaling and mitochondrial energy metabolism

  • Structural maturation:

    • KCNJ2 overexpression induces enlarged cell size and more elongated cell shape

    • Improves sarcomere organization

    • These changes can be visualized using KCNJ2 antibodies in combination with structural markers

  • Transcriptomic profile changes:

    • KCNJ2 overexpression alters the transcriptomic profile toward a more mature phenotype

    • Antibody-based techniques can be combined with transcriptomic analysis to correlate protein expression with gene expression changes

What are the solutions for common problems with KCNJ2 antibody applications?

ProblemPossible CausesSolutions
No signal in Western blotInsufficient protein loadingIncrease protein amount (30-50 μg)
Inefficient transferOptimize transfer conditions for membrane proteins
Inappropriate antibody dilutionTest dilution series (try 1:200 as starting point)
Multiple bandsNon-specific bindingInclude blocking peptide control
Cross-reactivity with other Kir channelsUse more specific antibody or validate with knockout
Protein degradationAdd fresh protease inhibitors to lysis buffer
No signal in immunostainingEpitope masking by fixationTry alternative fixation methods or use live cell staining
Insufficient permeabilizationOptimize detergent concentration and incubation time
Low expression levelUse signal amplification or more sensitive detection
High backgroundNon-specific bindingIncrease blocking time/concentration
Too high antibody concentrationDilute antibody (try 1:50 for immunocytochemistry)
Insufficient washingIncrease number and duration of wash steps

How to interpret discrepancies between different detection methods for KCNJ2?

When different detection methods yield inconsistent results:

  • Western blot vs. immunostaining discrepancies:

    • Western blot detects denatured protein while immunostaining typically detects native conformation

    • Epitope accessibility may differ between methods

    • Some antibodies work better for specific applications (check validation data)

  • Protein vs. mRNA level discrepancies:

    • Post-transcriptional regulation may cause differences

    • Protein stability and turnover affect steady-state levels

    • Consider time-course studies to address temporal dynamics

  • Functional vs. expression discrepancies:

    • Channel function depends on proper trafficking and post-translational modifications

    • High expression does not guarantee functional channels

    • Use combined approaches (e.g., patch-clamp with immunostaining of the same cell)

  • Species-specific discrepancies:

    • Epitope conservation varies across species

    • Validate antibodies separately for each species

    • Use multiple antibodies targeting different epitopes to confirm findings

How might new antibody technologies advance KCNJ2 research?

Emerging technologies promise to enhance KCNJ2 antibody applications:

  • Single-domain antibodies (nanobodies):

    • Smaller size allows better access to restricted epitopes

    • Potential for improved live-cell imaging of ion channels

    • May enable real-time tracking of channel dynamics

  • Intrabodies:

    • Antibody fragments that function within living cells

    • Could be used to track KCNJ2 trafficking in real-time

    • Potential for targeted modulation of channel function

  • Proximity proteomics:

    • Antibody-enzyme fusions for identifying proximal proteins

    • May reveal new KCNJ2 interaction partners

    • Could identify disease-specific alterations in the channel interactome

  • Super-resolution microscopy compatible tags:

    • Specific fluorophore conjugation for nanoscale imaging

    • May reveal channel nanodomain organization

    • Could identify changes in channel clustering in disease states

What are the emerging areas of KCNJ2 research where antibodies will play a crucial role?

Several research frontiers will benefit from advanced antibody applications:

  • Single-cell proteomics:

    • Quantifying channel expression variability within tissues

    • Correlating with electrophysiological heterogeneity

    • Antibodies will be essential for validation and calibration

  • Cardiac regenerative medicine:

    • Monitoring KCNJ2 expression during differentiation and maturation

    • Quality control of engineered cardiac tissues

    • KCNJ2 overexpression approaches to enhance cardiomyocyte maturation

  • Precision medicine for channelopathies:

    • Patient-specific iPSC-CM modeling of KCNJ2 mutations

    • Personalized drug screening based on channel expression and function

    • Antibodies will be crucial for phenotyping and validation

  • In vivo channel dynamics:

    • Development of antibody-based biosensors for real-time monitoring

    • Correlation of channel expression with cardiac arrhythmias

    • Integration with other physiological measurements

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.