The KCNV1 Antibody, HRP conjugated (Product Code: CSB-PA747493LB01HU) is a highly specific immunological tool designed for detecting potassium voltage-gated channel subfamily V member 1 (KCNV1) in human biological samples. This antibody is conjugated with horseradish peroxidase (HRP), enabling its use in enzyme-linked immunosorbent assays (ELISAs) and Western blotting for quantitative or qualitative analysis of KCNV1 expression.
Immunogen: Recombinant human KCNV1 protein (1-200AA).
Species Reactivity: Human.
Purification Method: Antigen affinity chromatography.
Conjugate: HRP for enhanced signal detection.
Storage: -20°C or -80°C; avoid repeated freeze-thaw cycles.
The HRP-conjugated KCNV1 antibody is optimized for sandwich ELISA protocols. When paired with a capture antibody (e.g., CSB-PA747493LB01HU), it enables precise quantification of KCNV1 in serum, plasma, or tissue homogenates. The assay employs a biotin-avidin amplification system, with a detection range tailored for physiological concentrations of KCNV1 .
Western blotting using this antibody involves SDS-PAGE separation of lysates, followed by membrane transfer and detection with chemiluminescent substrates. Optimal results are achieved with 1:500–1:1000 dilutions .
KCNV1 is implicated in modulating potassium channel activity, with roles in neuronal excitability and ion transport regulation. Dysregulation of KCNV1 has been linked to neurological disorders and cardiovascular diseases, including arrhythmias and neuropathic pain .
A 2022 study (PMC9253747) demonstrated that KCNV1 interacts with Kir7.1 channels in neuropathic pain models. Using Western blotting, researchers observed KCNV1-mediated inhibition of Kir7.1 activity, suggesting its role in pain signaling modulation .
In heart failure models, KCNA4 (a paralog of KCNV1) was found to regulate action potential duration. While KCNA4-specific antibodies were used, the findings highlight the broader relevance of potassium channel modulators like KCNV1 in arrhythmia prevention .
KCNV1, also known as potassium voltage-gated channel subfamily V member 1 or Kv8.1, represents a unique potassium channel subunit that does not form functional channels independently. Instead, it serves as a modulatory subunit that alters the properties of other potassium channels. KCNV1 functions by shifting the threshold for inactivation to more negative values and slowing the rate of inactivation when interacting with channels like KCNB1 and KCNB2. This modulation effectively changes the electrophysiological properties of these channels, potentially affecting neuronal excitability. Additionally, KCNV1 can downregulate channel activity of KCNB1, KCNB2, KCNC4, and KCND1, possibly through a mechanism involving trapping them in intracellular membranes . This regulatory role positions KCNV1 as an important modulator of cellular excitability rather than a direct mediator of ion flux across membranes.
Horseradish peroxidase (HRP) conjugation represents a critical modification to KCNV1 antibodies that enables direct detection through enzymatic reaction. The conjugation process covalently links HRP enzymes to the antibody structure, typically at sites that minimize interference with the antigen-binding region. This modification eliminates the need for secondary antibody steps in detection workflows, allowing for streamlined experimental protocols. The HRP enzyme catalyzes the oxidation of luminol in the presence of hydrogen peroxide, generating a chemiluminescent signal that can be detected and quantified . While the conjugation provides direct detection capabilities, researchers should be aware that it may potentially affect antibody affinity or specificity compared to unconjugated versions. Additionally, the molecular weight of the antibody increases with conjugation, which could impact penetration into fixed tissues or dense protein samples. Experimental validation is essential to confirm that the HRP conjugation doesn't compromise the antibody's ability to recognize KCNV1 specifically.
The KCNV1 Antibody, HRP conjugated has been primarily validated for enzyme-linked immunosorbent assay (ELISA) applications. According to product specifications, this conjugated antibody is optimized for detecting human KCNV1 in ELISA formats with recommended dilution ratios that should be empirically determined for each experimental system . The antibody has been generated using recombinant human potassium voltage-gated channel subfamily V member 1 protein (amino acids 1-200) as the immunogen, suggesting optimal reactivity against this region. Although primary validation has focused on ELISA applications, researchers may potentially adapt protocols for other applications such as immunohistochemistry, though additional optimization and validation would be required. When considering applications beyond the manufacturer's recommendations, preliminary testing with appropriate positive and negative controls is essential to establish reliability and specificity in the new experimental context.
Investigating KCNV1-KV7 interactions requires careful experimental design leveraging the KCNV1 antibody across multiple complementary approaches. First, consider transcript correlation analysis to identify potential physiologically relevant interactions. Single-cell RNA sequencing data has revealed strong correlations between KCNV1 and KCNQ2 (KV7.2) expression in hippocampal neurons, with Pearson's correlation coefficients for KCNV1 with KCNQ2 higher than 99% of all other transcripts . This provides a strong foundation for protein-level interaction studies. For direct interaction analysis, implement co-immunoprecipitation experiments using the KCNV1 antibody to pull down complexes, followed by immunoblotting for KV7 family members. To visualize interactions in native tissue, establish RNAScope protocols for simultaneous detection of both transcripts, focusing on regions with high expression such as the CA2 region of the hippocampus . For functional characterization, combine biochemical interaction data with electrophysiological recordings in heterologous expression systems or primary neurons to correlate complex formation with changes in channel properties. This multi-faceted approach provides complementary evidence supporting physiologically relevant interactions.
Rigorous validation of KCNV1 antibody specificity requires a comprehensive set of controls. First, implement tissue-specific positive and negative controls based on established expression patterns. Hippocampus and dorsal root ganglia samples serve as positive controls with known KCNV1 expression, while heart tissue can function as a negative control as it primarily expresses KCNV2 rather than KCNV1 . Second, perform peptide competition assays by pre-incubating the antibody with the immunizing peptide (amino acids 1-200 of KCNV1), which should eliminate specific signals while non-specific binding would remain. Third, establish genetic validation using KCNV1 knockdown or knockout models where specific signals should be abolished or significantly reduced. For Western blot applications, validate the specificity by confirming the molecular weight matches the predicted size for KCNV1, and include loading controls such as β-actin. Finally, compare results with alternative KCNV1 antibodies targeting different epitopes to establish concordance across detection methods, similar to validation approaches used for other potassium channel antibodies .
Characterizing the spatial distribution of KCNV1 in neuronal tissues requires employing multiple complementary techniques. First, implement immunohistochemistry using the KCNV1 antibody on fixed tissue sections, establishing appropriate antigen retrieval methods to overcome potential masking from fixation. For higher resolution analysis, employ single-molecule fluorescence RNA in-situ hybridization (RNAScope) which has successfully demonstrated KCNV1 expression patterns in hippocampal regions, with the highest signals observed in the CA2 region, followed by the dentate gyrus and CA1 region . For cellular and subcellular localization, combine the KCNV1 antibody with markers for specific neuronal compartments (such as MAP2 for dendrites or ankyrin-G for axon initial segments) using multi-color immunofluorescence and confocal microscopy. Super-resolution techniques like STORM or STED can further resolve subcellular localization at the nanometer scale. To correlate expression with functional properties, combine these imaging approaches with patch-clamp electrophysiology or calcium imaging in identified neurons expressing KCNV1, establishing structure-function relationships in specific neuronal populations.
Sample preparation methods must be tailored to the specific experimental context and the biochemical properties of KCNV1 as a membrane protein. For protein extraction from tissues expressing KCNV1 (such as hippocampus or dorsal root ganglia), employ specialized membrane protein extraction buffers containing appropriate detergents (0.5-1% Triton X-100, NP-40, or CHAPS) to efficiently solubilize KCNV1 while maintaining its native conformation. Include protease inhibitors to prevent degradation during processing. For immunohistochemistry applications, optimize fixation protocols, testing both paraformaldehyde (4%) and methanol fixation to determine which better preserves KCNV1 epitopes. Antigen retrieval methods (heat-induced or enzymatic) may be necessary to unmask epitopes, particularly in formalin-fixed tissues. For cell culture models expressing KCNV1, consider gentle permeabilization methods (0.1-0.2% Triton X-100 or saponin) to access intracellular epitopes while preserving membrane structures. When working with heterologous expression systems, timing of sample collection is critical, as overexpression may lead to retention in intracellular compartments rather than surface expression, potentially affecting detection patterns.
Optimizing signal-to-noise ratio for KCNV1 Antibody, HRP conjugated requires systematic adjustment of multiple experimental parameters. First, establish optimal antibody concentration through titration experiments, testing a range around the manufacturer's recommended dilution (0.1-0.5 μg/ml for similar antibodies) . Second, implement effective blocking procedures using 3-5% BSA or non-fat milk in PBS containing 0.05-0.1% Tween-20 to minimize non-specific binding, with extended blocking times (1-2 hours at room temperature) for challenging samples. Third, incorporate stringent washing steps between incubations, using PBS-T with optimized salt concentrations (150-300 mM NaCl) to reduce background while maintaining specific binding. Fourth, adjust substrate reaction conditions for optimal signal development - for chemiluminescence detection, monitor signal development in real-time to determine optimal exposure without background increase. For colorimetric detection, optimize substrate incubation time for maximum specific signal before background development. Finally, include chemical additives like polyethylene glycol (PEG) or dextran sulfate in antibody diluents to improve binding kinetics and reduce non-specific interactions, particularly in complex tissue samples.
When encountering detection challenges with KCNV1 Antibody, HRP conjugated, implement a systematic troubleshooting approach. For weak signals, first verify KCNV1 expression in your samples through RT-qPCR, as transcript levels vary significantly across tissues - hippocampus and dorsal root ganglia show high expression while heart tissue lacks KCNV1 expression . Increase antibody concentration incrementally (up to 3-5 times the recommended concentration) while monitoring background. For high background, enhance blocking by testing different blocking agents (BSA, casein, normal serum) and extending blocking times. Implement more stringent wash conditions with increased salt concentration or mild detergents. If detecting no signal, verify antibody activity with appropriate positive controls and check for potential interfering agents (particularly sodium azide which inactivates HRP). For membrane proteins like KCNV1, optimize antigen retrieval methods, testing both heat-induced (citrate or EDTA buffers at varying pH) and enzymatic approaches. If western blotting shows unexpected bands, adjust sample preparation to include stronger detergents for membrane protein extraction and optimize reducing conditions. For quantitative applications like ELISA, establish standard curves using recombinant KCNV1 protein to ensure measurements fall within the linear detection range.
KCNV1 exhibits distinct expression patterns across neuronal populations with significant regional and cell-type specificity. In the hippocampus, KCNV1 mRNA shows the highest expression in the CA2 region, followed by the dentate gyrus and CA1 region as demonstrated by single molecule fluorescence RNA in-situ hybridization . At the single-cell level, transcriptomic analysis has revealed KCNV1 presence across seven neuronal clusters in the hippocampus, with highest transcript counts in CA1-3 pyramidal neurons, dentate hilus, and dentate principal neurons. This expression profile partially overlaps with KCNQ2 (KV7.2), supporting potential functional interactions between these channel types. In dorsal root ganglia, KCNV1 is also expressed at levels comparable to KCNQ2, though interestingly, correlation analysis suggests stronger association with KV2 channel genes rather than KV7 channel genes in this tissue type . Notably, KCNV1 expression is absent in cardiac tissue, where instead KCNV2 (KV8.2) is expressed alongside KCNQ1 (KV7.1). This differential expression pattern across neuronal populations suggests specialized roles for KCNV1 in modulating neuronal excitability in a cell-type specific manner.
Detecting KCNV1 in fixed tissue samples requires carefully optimized immunohistochemistry protocols. Begin with appropriate tissue preparation: perfusion fixation with 4% paraformaldehyde for optimal preservation of membrane proteins, followed by cryoprotection in sucrose gradients before sectioning. For paraffin-embedded tissues, implement antigen retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) with heat treatment to unmask epitopes. Block sections with 5-10% normal serum from the species unrelated to the primary antibody host (rabbit in this case) and 0.3% Triton X-100 for permeabilization. Apply KCNV1 antibody at optimized concentration (starting with 2-5 μg/ml based on similar antibody recommendations) and incubate overnight at 4°C. For direct detection with HRP-conjugated antibodies, quench endogenous peroxidase activity with 0.3% hydrogen peroxide before antibody application. Perform stringent washing steps using PBS with 0.1% Tween-20. For chromogenic detection, develop with DAB substrate with careful timing to optimize signal-to-noise ratio. For fluorescence applications with HRP conjugates, use tyramide signal amplification systems for enhanced sensitivity. Include appropriate controls: primary antibody omission, concentration-matched isotype controls, and tissue with known KCNV1 expression patterns based on RNAScope findings .
Studying co-expression of KCNV1 with KV7 channels in hippocampal neurons requires multiple complementary approaches. First, implement dual fluorescence in situ hybridization (RNAScope) targeting both KCNV1 and KCNQ2 (KV7.2) transcripts simultaneously, focusing on regions like the CA2 region where KCNV1 shows highest expression . For protein-level analysis, employ dual-label immunohistochemistry using KCNV1 antibody alongside validated KV7.2 antibodies, with appropriate spectral separation of fluorophores. Confocal microscopy with z-stack acquisition enables three-dimensional reconstruction of expression patterns to determine subcellular co-localization. For higher resolution analysis, super-resolution microscopy techniques like STORM or STED can resolve co-localization at the nanometer scale. To assess physical interaction, implement proximity ligation assays (PLA) which generate fluorescent signals only when proteins are within 40nm of each other. For quantitative co-expression analysis across different hippocampal subregions, combine immunofluorescence with stereological sampling methods. Finally, correlate co-expression patterns with electrophysiological properties through patch-clamp recordings of identified neurons following post-hoc immunohistochemistry, establishing functional significance of KCNV1-KV7 co-expression patterns.
Investigating channel complex pharmacology involving KCNV1 requires sophisticated experimental approaches leveraging the HRP-conjugated antibody. First, establish heterologous expression systems co-expressing KCNV1 with established partners like KCNB1, KCNB2, or KCNQ2 based on transcriptomic correlation data from neuronal tissues . Implement ELISA-based binding assays using the HRP-conjugated antibody to screen compounds that may specifically disrupt or enhance complex formation, measuring changes in co-immunoprecipitation efficiency. Combine binding assays with electrophysiological characterization to correlate biochemical interaction with functional modulation of channel properties. Design high-throughput screening approaches using ELISA formats to identify compounds that alter the interaction between KCNV1 and KV7 channels but not KV7 homotetramers, potentially revealing compounds with selective pharmacological profiles. For compounds showing promising activity, perform dose-response studies combining surface expression assays (using the antibody to detect channel complexes) with patch-clamp electrophysiology to establish structure-function relationships. This integrated approach could identify novel therapeutic candidates targeting specific channel complexes rather than individual subunits, potentially offering improved selectivity profiles for neurological disorders.
Studying KCNV1's role in neurological disorders requires multi-faceted approaches combining the antibody with additional methodologies. First, analyze KCNV1 expression in post-mortem brain tissue from patients with relevant neurological disorders (such as epilepsy or neurodegenerative conditions) using immunohistochemistry with the KCNV1 antibody, comparing expression patterns and levels to matched control tissues. Given KCNV1's high expression in hippocampal CA2 region , focus on disorders affecting this region. Second, develop animal models with altered KCNV1 expression through genetic manipulation (knockout, knockdown, or overexpression) and assess resulting behavioral, electrophysiological, and biochemical phenotypes. Third, investigate if disease-associated mutations in KCNV1 interacting partners (such as KCNB1, KCNB2, or KCNQ2) affect their interaction with KCNV1 through co-immunoprecipitation and functional studies. Fourth, perform pharmacological studies using compounds known to modulate K+ channel function, assessing if their efficacy differs between wild-type and disease models in a KCNV1-dependent manner. Finally, evaluate correlations between KCNV1 genetic variants and disease susceptibility or progression through genomic studies, followed by functional validation of variant effects on channel complex formation and activity.
Studying KCNV1 trafficking and membrane localization requires sophisticated imaging approaches combined with biochemical techniques. First, implement surface biotinylation assays followed by pull-down with streptavidin beads and detection with KCNV1 antibody to quantify surface-expressed channels versus total cellular expression. For dynamic trafficking studies, employ pulse-chase approaches labeling surface proteins at different time points to track internalization rates. Combine the KCNV1 antibody with markers for different subcellular compartments (ER, Golgi, endosomes) in multi-color immunofluorescence to map the trafficking pathway. For live-cell imaging, develop a surface-labeling protocol using non-permeabilizing conditions with the antibody conjugated to pH-sensitive fluorophores to distinguish surface from internalized populations. Implement photobleaching techniques (FRAP) to measure lateral mobility of channel complexes in the membrane. For higher resolution analysis, combine the antibody with super-resolution microscopy techniques to visualize nanoscale clustering or localization to specialized membrane domains. To assess if KCNV1 indeed traps interacting channels in intracellular membranes as hypothesized , perform colocalization studies with markers for retention compartments like the ER or Golgi, comparing localization patterns in the presence versus absence of KCNV1 expression.