The rabbit anti-human TRPV2 polyclonal antibody is an IgG antibody produced in rabbits that specifically recognizes and binds to the human and mouse TRPV2 protein. Its immunogen is the recombinant human TRPV2 protein (1-117aa). Protein G purification of this TRPV2 antibody brought it a purity of up to 95%. The specificity and reliability of this TRPV2 antibody have been verified in ELISA, WB, IHC, and IF applications.
TRPV2 is primarily expressed in sensory neurons and regulates pain perception, body temperature, and cell survival. In particular, TRPV2 has been implicated in the regulation of cell proliferation, migration, and differentiation, as well as the modulation of synaptic plasticity in the nervous system. TRPV2 dysregulation is associated with various pathological conditions, including chronic pain, cancer, and neurodegenerative diseases.
The rabbit anti-human TRPV2 polyclonal antibody is an IgG antibody produced in rabbits that specifically recognizes and binds to the human and mouse TRPV2 protein. The immunogen is the recombinant human TRPV2 protein (1-117aa). This TRPV2 antibody has undergone Protein G purification, achieving a purity level of up to 95%. Its specificity and reliability have been validated in ELISA, WB, IHC, and IF applications.
TRPV2 is primarily expressed in sensory neurons and plays a crucial role in regulating pain perception, body temperature, and cell survival. Notably, TRPV2 has been implicated in the regulation of cell proliferation, migration, and differentiation, as well as the modulation of synaptic plasticity in the nervous system. Dysregulation of TRPV2 is associated with various pathological conditions, including chronic pain, cancer, and neurodegenerative diseases.
Applications : WB
Sample type: cells
Review: The rabbit polyclonal MITF antibody, rabbit polyclonal
When selecting TRPV2 antibodies, researchers should evaluate several key factors. First, consider the immunogen used for antibody generation - antibodies raised against full-length tetrameric TRPV2 often demonstrate superior specificity compared to those generated against synthetic peptides . Second, review validation data showing specificity against endogenous TRPV2, not just overexpressed protein. Third, select antibodies appropriate for your specific application (western blot, immunoprecipitation, or immunocytochemistry), as performance varies considerably between applications . For instance, monoclonal antibody 2D6 has shown excellent performance in western blot and immunoprecipitation, while 17A11 performs better in immunocytochemistry .
Comprehensive validation requires a multi-faceted approach. Begin by testing antibody recognition against recombinant full-length TRPV2 alongside N-terminal and C-terminal fragments to map binding regions . Conduct siRNA knockdown experiments in cells expressing endogenous TRPV2, such as F11 cells, to confirm signal reduction following TRPV2 depletion . Test for cross-reactivity with closely related proteins, particularly TRPV1, which shares approximately 50% sequence identity with TRPV2 . For rigorous validation, compare results across antibodies targeting different epitopes and include appropriate negative controls. Commercial polyclonal antibodies against TRPV2 C-terminal peptides often show non-specific binding patterns that are insensitive to TRPV2 knockdown, highlighting the importance of proper validation .
The optimal methodology varies by application. For western blot detection, monoclonal antibodies targeting the C-terminus (like 2D6) demonstrate superior specificity compared to commercial polyclonal antibodies . For immunoprecipitation of endogenous TRPV2 from tissue samples, 2D6 antibody has successfully precipitated TRPV2 from mouse brain and heart when other antibodies failed . For immunocytochemistry, 17A11 shows specific recognition of TRPV2 with minimal cross-reactivity . When using immunocytochemistry, optimal results are achieved with 4% paraformaldehyde fixation followed by permeabilization with 0.3% Triton X-100 in PBS containing normal serum .
Detecting endogenous TRPV2 presents significant challenges. Endogenous TRPV2 protein is expressed at very low levels in most tissues, requiring highly sensitive detection methods . Non-specific antibody binding becomes proportionally more problematic with low-abundance targets. Tissue lysate analysis reveals minimal TRPV2 immunoreactivity in whole tissue extracts, suggesting low endogenous expression levels even in tissues with documented TRPV2 mRNA . Immunoprecipitation before western blotting can enhance detection sensitivity. When investigating TRPV2 in specific tissues, focus on those with higher expression levels, such as dorsal root ganglia, brain, and heart . Always include appropriate controls, particularly siRNA knockdown validation, to confirm specificity of detected signals.
The standard protocol involves several key steps. Begin by homogenizing tissue in lysis buffer using a dounce homogenizer (approximately 10 strokes) . Incubate homogenates on ice for 30 minutes, then clear by centrifugation at 20,000×g for 20 minutes followed by 100,000×g for 30 minutes to remove unbroken cells and insoluble membrane fragments . Pre-clear the supernatant with protein A/G agarose beads, then incubate pre-cleared lysate (approximately 2.5 mg) with 10 μg anti-TRPV2 antibody for 2 hours at 4°C . Add 50 μl protein A/G agarose for 2 hours at 4°C to capture TRPV2 antibody complexes . Wash beads three times in lysis buffer and elute proteins with Laemmli sample buffer boiled at 95°C for 5 minutes . Analyze immunoprecipitation by western blot with appropriately labeled secondary antibodies.
Distinguishing membrane-associated from intracellular TRPV2 requires careful methodological approaches. Current evidence suggests that TRPV2 primarily resides in intracellular membranes rather than at the plasma membrane . To accurately assess localization, perform selective plasma membrane labeling using non-permeabilized cells, followed by total TRPV2 staining after permeabilization. Use membrane markers for co-localization studies and employ confocal microscopy with Z-stack imaging to visualize membrane versus intracellular distribution. For heterologously expressed TRPV2, epitope tagging (such as 1D4-tagging) provides a reliable reference for subcellular localization . When studying trafficking, employ both immunocytochemistry and complementary approaches like cell surface biotinylation assays, which can provide quantitative assessment of plasma membrane expression .
TRPV2 plays a critical role in B cell activation through multiple mechanisms. Research demonstrates that TRPV2 is highly expressed in B cells and contributes significantly to B cell immunological synapse formation and activation . Upon antigen stimulation, TRPV2 mediates calcium influx, influencing membrane potential depolarization and promoting cytoskeleton remodeling within the immunological synapse . Physiologically, TRPV2 expression levels positively correlate with influenza-specific antibody production . This correlation has age-related implications, as TRPV2 expression is lower in newborns and seniors, potentially contributing to reduced antibody responses in these populations .
To investigate TRPV2's role in B cell immunological synapse formation, researchers should employ multiple complementary approaches. Live-cell calcium imaging using fluorescent indicators can monitor TRPV2-mediated calcium influx during synapse formation . Confocal or super-resolution microscopy with validated antibodies allows visualization of TRPV2 localization within the synapse . B cell-specific TRPV2 knockout models provide powerful tools to assess functional consequences of TRPV2 deletion, as mice with B cell-specific TRPV2 deficiency display impaired antibody responses following immunization . Domain-specific mutations can identify regions critical for synapse formation, with particular attention to the pore and N-terminal domains which are crucial for gating cation permeation and executing mechanosensation in B cells upon antigen stimulation .
The trafficking behavior of TRPV2 has been controversial in scientific literature. While early studies suggested that insulin-like growth factor 1 (IGF-1) increases TRPV2 trafficking to the plasma membrane, more recent research using validated monoclonal antibodies has challenged this view . Both cell surface biotinylation assays and immunocytochemistry indicate that IGF-1 has little to no effect on TRPV2 surface expression in cells transiently expressing TRPV2 . Similarly, in F11 cells expressing endogenous TRPV2, little to no TRPV2 was detected at the cell surface regardless of IGF-1 treatment . Current evidence suggests that TRPV2 primarily resides in intracellular membranes and its subcellular distribution is largely insensitive to IGF-1 treatment .
Resolving conflicting data about TRPV2 trafficking requires rigorous experimental design and multiple complementary approaches. Cell surface biotinylation assays using membrane-impermeable biotin reagents followed by streptavidin pulldown and western blotting provide quantitative assessment of surface expression . These should be complemented with immunocytochemistry in both permeabilized and non-permeabilized cells to distinguish surface from intracellular protein. When studying IGF-1 effects, carefully control treatment conditions and include appropriate time courses. The controversy surrounding IGF-1-induced TRPV2 translocation highlights the critical importance of antibody validation - earlier studies using incompletely characterized antibodies may have yielded misleading results . While PI3 kinase signaling modulates TRPV2 activity, its effects on regulated insertion of TRPV2 into the plasma membrane remain contentious .
TRPV2 expression shows significant alterations in several disease states with important pathophysiological implications. In systemic lupus erythematosus (SLE), researchers have established a positive correlation between TRPV2 expression levels and clinical disease manifestations in both adult and pediatric patients . This suggests TRPV2 may contribute to autoimmune pathology through enhanced B cell activation. Age-related differences in TRPV2 expression have also been observed, with lower levels in newborns and seniors correlating with reduced influenza-specific antibody production . Beyond immune-related conditions, altered TRPV2 expression and distribution have been implicated in muscular dystrophy, cardiomyopathy, and certain cancers , suggesting that monitoring TRPV2 expression may provide insights into disease mechanisms or serve as a potential biomarker.
Investigating TRPV2's role in autoimmune diseases requires sophisticated methodological approaches. For clinical studies, compare TRPV2 expression levels in immune cells from patients versus healthy controls using validated antibodies and flow cytometry or western blotting . Generate conditional knockout models (B cell-specific TRPV2 knockout mice) and assess autoantibody production and disease manifestations in autoimmune-prone genetic backgrounds . Ex vivo functional assays with isolated B cells from patients and controls can compare calcium signaling, immunological synapse formation, and antibody production. The mechanistic connection between TRPV2 and B cell hyperactivity in SLE represents a particularly promising research direction, as the pore and N-terminal domains of TRPV2 contribute to membrane potential depolarization and cytoskeleton remodeling within the B cell immunological synapse , processes that may be dysregulated in autoimmunity.
Cross-reactivity assessment is essential for TRPV2 antibody validation. Conduct comparative western blot analysis using recombinant proteins or cell lysates expressing individual TRP family members, particularly the closely related TRPV1, which shares approximately 50% sequence identity with TRPV2 . Perform immunocytochemistry in cells expressing either TRPV2 or other TRP channels to evaluate antibody specificity . Preabsorption studies, where the antibody is pre-incubated with recombinant target or related proteins prior to use, can reveal cross-reactivity. Given that most available polyclonal TRPV2 antibodies were generated against the C-terminus (where sequence divergence from other TRPV subfamily members is greatest), it is surprising that many commercial antibodies still show significant cross-reactivity .
To minimize TRPV1 cross-reactivity, select antibodies raised against the C-terminal region of TRPV2, which shows greater sequence divergence from TRPV1 . Validate using both TRPV1 and TRPV2 expressed in the same system - for example, monoclonal antibody 2D6 recognizes TRPV2 but not TRPV1 in HeLa cells . Generate epitope-specific antibodies targeting unique sequences in TRPV2 with minimal homology to TRPV1. Consider using monoclonal antibodies raised against full-length tetrameric TRPV2, which have shown superior specificity compared to polyclonal antibodies raised against synthetic peptides . When studying tissues expressing both channels, include appropriate controls to distinguish signals. The development of monoclonal antibodies against full-length tetrameric TRPV2 represents an important methodological advance that may be applicable for generating antibodies against other TRP channels with unclear functions .
Several methodological innovations have significantly improved TRPV2 detection. The development of monoclonal antibodies against full-length tetrameric TRPV2 (rather than peptide fragments) has dramatically enhanced specificity and reduced false positives . These antibodies show superior performance in recognizing native TRPV2 conformations compared to antibodies raised against synthetic or recombinant linear peptides . Application-specific antibody selection (using 2D6 for western blot/immunoprecipitation and 17A11 for immunocytochemistry) optimizes detection across different experimental contexts . IR-dye-labeled secondary antibodies enhance detection sensitivity for low-abundance endogenous TRPV2 . These advances have clarified contradictory findings regarding TRPV2 trafficking and localization that resulted from inadequately validated antibodies in earlier studies .
Single-cell approaches offer powerful tools for elucidating TRPV2 function. Single-cell calcium imaging combined with patch-clamp electrophysiology can directly measure TRPV2-mediated cation currents during cellular activation . In B cells specifically, single-cell imaging can visualize TRPV2 dynamics during immunological synapse formation and correlate channel localization with calcium signaling . Single-cell RNA sequencing can identify cell populations with varying TRPV2 expression levels and correlate expression with functional phenotypes. For B cells, correlating single-cell TRPV2 expression with antibody production capacity could reveal important functional relationships . These approaches are particularly valuable given the heterogeneity in TRPV2 expression across different cell types and developmental stages, and the correlation between expression levels and functional outcomes like antibody production .
Essential controls for TRPV2 antibody experiments include several key components. For western blot, include positive controls using recombinant TRPV2 protein or lysates from cells overexpressing TRPV2 . Negative controls should use lysates from cells with confirmed absence of TRPV2 expression or siRNA-mediated TRPV2 knockdown . For immunoprecipitation, include a non-specific IgG control to identify non-specific binding . In immunocytochemistry, use cells expressing epitope-tagged TRPV2 (such as 1D4-tagged TRPV2) as positive controls, with the epitope antibody providing a reference for correct subcellular localization . Secondary antibody-only controls identify non-specific binding. Whenever possible, compare results using at least two validated antibodies targeting different TRPV2 epitopes to confirm findings .
siRNA knockdown experiments provide powerful validation of antibody specificity. Select cells with confirmed endogenous TRPV2 expression, such as F11 cells derived from dorsal root ganglion neurons . Design siRNAs targeting conserved regions of TRPV2 and use multiple siRNAs to confirm results. Include non-targeting siRNA as a negative control. Allow sufficient time for protein turnover (typically 48-72 hours post-transfection) before analysis. Quantify band intensity between TRPV2 siRNA and non-targeting control siRNA treated samples - effective knockdown should reduce signal by at least 50-70% . Confirm that levels of related proteins remain unchanged to rule out off-target effects. When properly executed, siRNA knockdown provides compelling evidence for antibody specificity, as demonstrated with monoclonal antibody 2D6, which showed a nearly 3-fold reduction in band intensity at the molecular weight corresponding to TRPV2 in F11 cells treated with TRPV2 siRNA .
Several promising research directions emerge from recent TRPV2 findings. Investigation of TRPV2's role in B cell activation and antibody production offers opportunities for understanding immune response regulation and potential therapeutic interventions . Exploring the correlation between TRPV2 expression and autoimmune disease severity, particularly in SLE, may yield new biomarkers or treatment targets . Age-related differences in TRPV2 expression and their impact on immune responses, especially in newborns and seniors, represent an important area for vaccine research . The role of TRPV2 in disease states including muscular dystrophy, cardiomyopathy, and cancer requires further investigation . Development of more specific pharmacological modulators of TRPV2 would greatly advance the field, as current tools lack specificity .
Structural biology approaches offer significant potential for advancing TRPV2 antibody development. Cryo-electron microscopy of TRPV2 in different conformational states can identify exposed epitopes ideal for antibody targeting. Structure-guided epitope selection focusing on regions with minimal conservation across TRP channels would enhance specificity. Computational modeling of antibody-TRPV2 interactions could predict cross-reactivity and guide antibody engineering. The successful generation of monoclonal antibodies against full-length tetrameric TRPV2 demonstrates the value of targeting native protein conformations rather than peptide fragments . Future efforts might employ phage display technologies with structural information to develop conformation-specific antibodies that distinguish between active and inactive TRPV2 states. These approaches could yield next-generation antibodies with enhanced specificity and application-specific properties.