Detects HPCAL4 at 22 kDa in brain lysates (human, mouse, rat) .
Validated in cell lines (HEK-293T, U-251MG) and tissues (brain, liver) .
Localizes HPCAL4 in human medulloblastoma and mouse spinal cord neurons .
Protocols recommend antigen retrieval with TE buffer (pH 9.0) .
Calcium Signaling: Regulates rhodopsin phosphorylation and synaptic plasticity .
Pain and Itch Processing: Mouse studies show mild thermal sensitivity deficits in Hpcal4 knockouts but no major impact on acute pain or itch .
Neurological Disorders: Linked to excitatory interneuron function in spinal cord pathways .
HPCAL4 (hippocalcin-like 4) is a calcium-binding protein belonging to the Recoverin family. In humans, the canonical protein consists of 191 amino acid residues with a molecular mass of approximately 22.2 kDa. This protein is primarily involved in calcium-dependent regulation of rhodopsin phosphorylation. HPCAL4 is also known by the synonyms HLP4 and hippocalcin-like protein 4. Gene orthologs have been identified across multiple species including mouse, rat, bovine, frog, chimpanzee, and chicken . Research indicates that HPCAL4 is heavily expressed in the central nervous system, particularly in neurons of the superficial dorsal horn of the spinal cord (laminae I-II) .
HPCAL4 antibodies are versatile immunological tools employed across multiple experimental procedures. Western blotting represents the most common application, where these antibodies effectively detect the 22 kDa HPCAL4 protein in brain tissue samples from humans, mice, and rats . Beyond Western blotting, HPCAL4 antibodies demonstrate utility in immunoprecipitation protocols, particularly with fetal human brain tissue. Additionally, these antibodies can be applied in immunohistochemistry applications, with specific effectiveness in human medulloblastoma tissue samples . Some HPCAL4 antibodies also perform well in ELISA assays, providing researchers with multiple experimental approaches for investigating this calcium-binding protein .
When investigating HPCAL4 expression patterns, researchers should anticipate pronounced expression in neural tissues, particularly the central nervous system. Within the spinal cord, in situ hybridization studies demonstrate that HPCAL4 expression is concentrated in the substantia gelatinosa (laminae I-II), which represents a critical area for nociceptive processing . Antibody-based detection methods have successfully identified HPCAL4 in human brain tissue (both adult and fetal), mouse brain tissue, and rat brain tissue samples . Single-cell transcriptome analysis has revealed that certain subtypes of unmyelinated sensory neurons, specifically peptidergic and tyrosine hydroxylase-expressing subpopulations, also express HPCAL4 . This distinct neuroanatomical distribution pattern suggests specialized functions in sensory processing circuits.
For optimal Western blot detection of HPCAL4, researchers should implement a carefully calibrated protocol. Begin with appropriate sample preparation, focusing on neural tissues including human, mouse, or rat brain samples, which demonstrate reliable HPCAL4 expression . When selecting antibody dilutions, empirical testing suggests that a range of 1:1000 to 1:4000 provides optimal signal-to-noise ratios for most HPCAL4 antibodies . The expected molecular weight band for HPCAL4 is approximately 22 kDa, consistent with its calculated molecular mass. For enhanced specificity, researchers should consider optimization of blocking conditions, incubation times, and washing steps. When analyzing results, validation through positive controls (brain tissue lysates) is essential, as is confirmation of specificity through knockout controls when possible . If weak signals are encountered, enrichment through immunoprecipitation prior to Western blotting may enhance detection of low-abundance HPCAL4 protein.
Implementation of rigorous controls is essential for generating reliable data with HPCAL4 antibodies. Positive tissue controls should include human, mouse, or rat brain samples, which consistently demonstrate HPCAL4 expression across multiple studies . When available, researchers should utilize genetically modified models (such as HPCAL4 knockout mice) as negative controls to definitively validate antibody specificity . In the absence of genetic models, antibody validation can be performed through complementary approaches including pre-absorption controls with immunizing peptides, comparison of staining patterns across multiple antibodies targeting different HPCAL4 epitopes, and correlation with mRNA expression data through in situ hybridization . For quantitative applications, researchers should implement calibration standards and ensure linear detection ranges. Additionally, when comparing HPCAL4 expression across experimental conditions, careful normalization to appropriate housekeeping proteins or internal controls is essential to account for variations in sample loading and processing.
To investigate HPCAL4's role in pain processing, researchers should implement a multifaceted experimental approach. Genetic manipulation techniques provide valuable insights, as demonstrated by studies utilizing global Hpcal4 knockout mice that revealed mild but significant thermal deficits in the Hargreaves and hotplate tests . For more targeted investigations, conditional knockout strategies employing Cre-loxP systems would allow tissue-specific deletion of Hpcal4, enabling discrimination between peripheral and central contributions to observed phenotypes. Complementary pharmacological approaches using calcium signaling modulators could further elucidate HPCAL4's functional mechanisms. Researchers should employ comprehensive behavioral testing batteries that assess multiple pain modalities, including mechanical sensitivity (von Frey filaments), thermal responsiveness (Hargreaves, hotplate, tail immersion tests), and chemical nociception (capsaicin, formalin) . Importantly, both acute and persistent pain models should be evaluated, as Hpcal4 knockout effects may be context-dependent. Additionally, investigating sex-specific effects is crucial, as previous research has identified sex-dependent phenotypes in thermal sensitivity tests with Hpcal4 knockout mice .
Investigation of HPCAL4's calcium-binding properties requires sophisticated biochemical and molecular approaches. Researchers should consider employing calcium-binding assays using purified recombinant HPCAL4 protein to determine binding kinetics, stoichiometry, and calcium concentration dependencies. As a member of the Recoverin family, HPCAL4 is suspected to regulate rhodopsin phosphorylation in a calcium-dependent manner , suggesting functional analyses should include phosphorylation assays in the presence of varying calcium concentrations. Advanced structural biology techniques including X-ray crystallography or cryo-electron microscopy would provide valuable insights into conformational changes associated with calcium binding. For cellular investigations, researchers might implement calcium imaging in combination with HPCAL4 overexpression or knockdown to assess how this protein modulates intracellular calcium dynamics. To identify downstream signaling partners, co-immunoprecipitation followed by mass spectrometry analysis represents a powerful unbiased approach. Additionally, proximity labeling techniques such as BioID or APEX could identify proteins that transiently interact with HPCAL4 in a calcium-dependent manner, potentially revealing novel signaling pathways.
Investigating alterations in HPCAL4 expression under pathological conditions requires careful experimental design. Researchers should consider implementing animal models of neurological disorders, particularly those involving calcium dysregulation or sensory processing deficits. The demonstrated downregulation of HPCAL4 in TR4 mutant mice suggests that transcriptional regulation of this gene responds to pathophysiological changes. Quantitative approaches including qPCR, Western blotting, and immunohistochemistry with carefully validated antibodies can assess expression changes at mRNA and protein levels. For human studies, examination of postmortem tissue samples from individuals with relevant neurological conditions, compared with appropriate controls, may reveal disease-associated alterations in HPCAL4 expression. Researchers might also consider analyzing publicly available transcriptomic datasets from conditions including neuropathic pain, inflammatory disorders, or neurodegenerative diseases to identify potential correlations with HPCAL4 expression. Single-cell RNA sequencing approaches would provide particularly valuable insights into cell type-specific alterations in HPCAL4 expression under pathological conditions. Understanding these expression patterns could reveal potential therapeutic targets for conditions involving sensory processing dysfunction.
Addressing inconsistent HPCAL4 antibody performance requires systematic troubleshooting approaches. First, researchers should carefully evaluate antibody specifications, including the immunogen used for production, as antibodies generated against different epitopes may perform differently across applications . For Western blotting inconsistencies, optimization of sample preparation is critical—HPCAL4 being a calcium-binding protein might demonstrate altered migration patterns under different buffer conditions. Researchers should test multiple protein extraction methods, focusing on those designed for membrane-associated proteins. For immunohistochemistry applications, comparison of different antigen retrieval methods (TE buffer pH 9.0 versus citrate buffer pH 6.0) might resolve detection issues . Additionally, sensitivity can be enhanced through signal amplification systems such as tyramide signal amplification. When inconsistencies persist, researchers should consider testing multiple antibodies targeting different HPCAL4 epitopes to determine whether observed patterns are antibody-specific or represent true biological variation. Complementary detection methods, such as mRNA localization through in situ hybridization, can provide independent validation of expression patterns .
The interpretation of subtle phenotypes in HPCAL4 knockout models requires careful consideration of several factors. Researchers should recognize that mild phenotypes, such as the thermal sensitivity deficits observed in Hpcal4 knockout mice , might reflect functional redundancy among calcium-binding proteins or compensatory mechanisms activated during development. To address this possibility, investigations employing acute knockdown approaches (e.g., siRNA, antisense oligonucleotides) or inducible knockout systems might reveal more pronounced phenotypes by circumventing developmental compensation. When analyzing behavioral data, sophisticated statistical approaches beyond simple group comparisons might uncover subtle patterns in response distributions or temporal dynamics. Additionally, researchers should consider that HPCAL4's functional role might be context-dependent, becoming more prominent under specific pathological conditions rather than in baseline states. Combinatorial approaches targeting multiple calcium-binding proteins simultaneously might reveal synergistic interactions that are not apparent in single-gene knockout models. Finally, the observation that sex-dependent effects exist in certain thermal sensitivity assays highlights the importance of analyzing male and female subjects separately rather than pooling data, which might obscure important biological differences.
Designing robust experiments to investigate HPCAL4's calcium-dependent functions requires careful consideration of multiple factors. Researchers should implement precise control of calcium concentrations, potentially using calcium chelators (EGTA, BAPTA) to establish calcium-free conditions and calcium ionophores to experimentally manipulate intracellular calcium levels. Given HPCAL4's suspected involvement in rhodopsin phosphorylation regulation , phosphorylation assays should be conducted across a range of physiologically relevant calcium concentrations (typically 100 nM to 10 μM). When performing cellular studies, selection of appropriate model systems is critical—given HPCAL4's expression pattern, primary neuronal cultures or neuronal cell lines would provide relevant biological context . For protein interaction studies, researchers should be aware that calcium-dependent interactions may be transient or low-affinity, requiring specialized approaches such as chemical crosslinking or proximity labeling rather than traditional co-immunoprecipitation. When designing mutational studies, careful consideration should be given to the calcium-binding EF-hand domains within HPCAL4, with targeted mutations that disrupt calcium coordination providing valuable insights into function. Finally, researchers should consider the spatial and temporal dynamics of calcium signaling, potentially employing live-cell imaging with genetically encoded calcium indicators alongside fluorescently tagged HPCAL4 to visualize dynamic interactions in real time.
HPCAL4 antibodies can serve as valuable models in therapeutic antibody development research. Recent advancements in antibody engineering, as demonstrated in studies focusing on enhancing thermostability and affinity, provide a methodological framework applicable to HPCAL4 antibodies . Researchers can employ deep learning models like DeepAb to predict antibody structures directly from sequence data, then combine this computational approach with experimental deep mutational scanning (DMS) to design optimized variants . This integrated strategy has shown remarkable success, with some engineered antibodies demonstrating significantly increased affinity (5-21 fold) and enhanced thermostability (>2.5°C increase in melting temperature) . When applied to HPCAL4 antibodies, researchers should systematically evaluate thermal stability parameters (Tonset, Tm, Tagg), binding affinity (KD), and developability characteristics (non-specific binding, aggregation propensity, self-association) of engineered variants compared to parental antibodies . Importantly, this approach allows for antibody optimization without requiring crystal structures of antibody-antigen complexes, which are often unavailable for novel targets like HPCAL4 .
Current evidence suggests HPCAL4 has specialized functions in distinct sensory neuron populations. Single-cell transcriptome analyses have revealed HPCAL4 expression in specific subtypes of unmyelinated sensory neurons, particularly those expressing peptides and tyrosine hydroxylase . This selective expression pattern suggests potential roles in modulating sensory transduction or neurotransmission in these neuronal subpopulations. The mild thermal sensitivity phenotypes observed in Hpcal4 knockout mice align with potential functions in heat-sensitive neurons, though the subtlety of these effects suggests complex interactions with other regulatory molecules. Researchers investigating HPCAL4's subtype-specific functions should consider implementing targeted approaches such as conditional knockout strategies restricted to defined neuronal populations (e.g., using TH-Cre or peptidergic neuron-specific Cre lines). Additionally, ex vivo electrophysiological recordings from identified sensory neurons in Hpcal4 knockout versus wild-type mice could provide direct evidence of altered firing properties or calcium handling. Calcium imaging experiments in identified neuronal subtypes would also offer insights into how HPCAL4 might regulate calcium transients during sensory activation. Integration of these approaches with behavioral testing would establish connections between molecular mechanisms and sensory function.
The relationship between HPCAL4 and TR4 (testicular receptor 4) represents an intriguing area for further investigation, as HPCAL4 expression is significantly downregulated in TR4 mutant mice . To elucidate this regulatory relationship, researchers should implement complementary molecular approaches. Chromatin immunoprecipitation (ChIP) assays could determine whether TR4, as a nuclear receptor, directly binds to the HPCAL4 promoter region to regulate its transcription. Reporter gene assays using HPCAL4 promoter constructs with wild-type or mutated potential TR4 binding sites would provide functional validation of direct regulation. Conversely, investigation of potential indirect regulatory mechanisms might focus on intermediate transcription factors or signaling pathways. Single-cell transcriptomic analyses comparing wild-type and TR4 mutant spinal cord neurons would identify co-regulated gene networks, potentially revealing the broader context of HPCAL4 regulation . Developmental studies examining the temporal dynamics of TR4 and HPCAL4 expression during neuronal differentiation could provide insights into critical periods of regulation. Additionally, rescue experiments introducing HPCAL4 expression in TR4 mutant backgrounds would determine whether HPCAL4 downregulation contributes to the observed pain and itch processing deficits in these mutant mice . This multifaceted approach would establish whether HPCAL4 represents a key downstream effector of TR4 signaling in sensory processing circuits.