Protocol: MAB70441 detects CBLN2 in paraffin-embedded human brain sections (medulla) at 15 µg/mL, with localization to neuronal cell bodies .
Staining: Anti-mouse HRP-DAB kits yield brown chromogenic signals, counterstained with hematoxylin .
Cerebellar Synapses: CBLN2 associates with CBLN1 to stabilize synapses, though Cbln2-null mice lack striatal synaptic defects seen in Cbln1-null models .
Behavioral Roles: Conditional Cbln2 deletion in the medial habenula impairs synaptic transmission in interpeduncular neurons, reducing spatial learning in mice .
Pulmonary Hypertension: CBLN2 promotes endothelial-mesenchymal transition via NF-κB/HIF-1α/Twist1 pathways .
Cancer: Overexpression inhibits colorectal cancer progression and enhances immunotherapy responses .
CBLN2 (Cerebellin 2 Precursor) is a 24 kDa cytoplasmic, membrane, and secreted glycoprotein that forms intracellular and extracellular homodimers and heterodimers . It belongs to the cerebellin family of proteins that regulate synapse development and function . CBLN2 exhibits functional redundancy with Cbln1 in the cerebellum, though it demonstrates distinct properties in thalamic neurons . The protein plays a significant role in neural development, particularly in the prefrontal cortex where it has been linked to increased postsynaptic structures and synapse formation, potentially contributing to the evolutionary development of higher-order cognitive functions in primates . Understanding CBLN2 function can provide insights into neuronal connectivity patterns and synaptic regulation mechanisms relevant to both normal brain development and neurological disorders.
CBLN2 antibodies have multiple validated applications in laboratory research. Based on available data, these antibodies can be successfully employed in Western Blotting (WB) for protein detection and quantification in tissue or cell lysates . They are also effective in Immunohistochemistry on paraffin-embedded sections (IHC-p), allowing visualization of CBLN2 distribution in fixed tissue samples . Additional validated applications include Enzyme-Linked Immunosorbent Assay (ELISA) for quantitative measurement in solution, Immunocytochemistry (ICC) for cellular localization studies, and various Immunofluorescence (IF) approaches for high-resolution imaging of CBLN2 distribution . When selecting a CBLN2 antibody for your experiment, it's essential to verify that the specific antibody has been validated for your intended application and species of interest, as reactivity patterns vary between antibody products.
Confirming antibody specificity is crucial for reliable CBLN2 research. A multi-method validation approach is recommended, beginning with Western blot analysis using positive and negative control samples. For CBLN2, this can include lysates from tissues known to express the protein (positive control) versus knockout tissues or cells (negative control) . Immunoblotting of lysates from cells transfected with recombinant vectors expressing various cerebellin family members (CBLN1-4) can distinguish cross-reactivity within this protein family . Immunohistochemistry patterns should match known expression profiles of CBLN2 across brain regions. Additionally, peptide competition assays can verify epitope specificity - preincubation of the antibody with excess immunizing peptide should abolish signal in subsequent assays . For advanced validation, researchers have employed affinity column purification with peptide antigens to enhance antibody specificity, as demonstrated in previous work using a synthetic peptide corresponding to amino acids 88-104 of mouse Cbln2 (NP_766221) .
When performing Western blotting with CBLN2 antibodies, several technical considerations are essential for successful detection. First, ensure proper sample preparation - CBLN2 is a 24 kDa protein that can form dimers, so reducing conditions are typically required for consistent band detection . Select an appropriate antibody targeting either the N-terminal or C-terminal region depending on your research question, as different epitopes may reveal distinct protein forms or fragments . For example, antibodies targeting the C-terminal region (AA 179-207) have proven effective in detecting CBLN2 in human and mouse samples . When optimizing protein loading, consider that CBLN2 expression varies significantly between tissues and cell types, with notable expression in neural tissues. Blocking conditions should be optimized to minimize background signal while maintaining specific binding. When comparing CBLN2 expression between samples (e.g., between chemoresistant and chemosensitive cell lines), ensure equal protein loading using appropriate housekeeping controls . For challenging detections, signal enhancement systems may be necessary, particularly when examining endogenous CBLN2 in tissues with lower expression levels.
Differentiating between CBLN family members presents a significant challenge due to structural similarities and potential functional redundancy. Researchers should employ antibodies targeting unique, non-conserved regions of CBLN2 to minimize cross-reactivity with CBLN1, CBLN3, and CBLN4 . The epitope selection is critical - antibodies directed against the region N-terminal to the C1q motif in CBLN2 (such as those targeting amino acids 88-104 in mouse Cbln2) have demonstrated high specificity . For ultimate validation, perform parallel experiments using tissues or cells from knockout models lacking specific CBLN family members. When conducting co-expression studies, use antibodies raised in different host species to allow simultaneous detection of multiple CBLN proteins. Western blot analysis can help distinguish family members based on subtle size differences, though this requires high-resolution gel systems. If cross-reactivity remains a concern, consider complementary approaches such as RNA-based detection methods (RT-qPCR, in situ hybridization) targeting unique nucleotide sequences, which can circumvent antibody specificity limitations. Mass spectrometry-based proteomic analysis provides another orthogonal validation approach, identifying unique peptide signatures for each family member.
When faced with contradictory results using different CBLN2 antibodies, systematic troubleshooting is essential. First, compare the epitope specificities of each antibody - differences may reflect detection of distinct CBLN2 domains, post-translational modifications, or protein conformations . Create a validation matrix testing each antibody against recombinant CBLN2 proteins with defined modifications or truncations to map epitope accessibility under various conditions. Examine whether discrepancies correlate with the antibody's production method (polyclonal vs. monoclonal) or immunogen design (synthetic peptide vs. full protein) . For polyclonal antibodies like the affinity-purified rabbit anti-CBLN2 described in the literature, batch-to-batch variability may contribute to inconsistent results . Consider whether sample preparation methods differentially affect epitope availability - factors such as fixation protocol, antigen retrieval method, or protein denaturation conditions can dramatically influence antibody binding. Cross-validate findings using complementary detection methods independent of antibodies, such as mass spectrometry or CRISPR-based tagging of endogenous CBLN2. Finally, biological context matters - CBLN2 undergoes complex processing and trafficking, potentially resulting in different subcellular localization patterns that may be preferentially detected by different antibodies, as evidenced by studies showing CBLN2 in both cytoplasmic and vesicle-like structures .
To investigate CBLN2's role in synaptic development, researchers can employ multi-modal antibody-based approaches. Immunofluorescence co-localization studies using CBLN2 antibodies alongside pre- and post-synaptic markers (such as synaptophysin and PSD-95/DLG4) can quantify changes in synaptic density associated with CBLN2 expression . High-resolution techniques like super-resolution microscopy or electron microscopy with immunogold labeling provide nanoscale insights into CBLN2's precise localization at synapses. For functional studies, researchers can combine immunocytochemistry with electrophysiological recordings to correlate CBLN2 expression patterns with synaptic strength. Time-course experiments during development are particularly informative, as CBLN2 expression shows temporal regulation that impacts synaptogenesis, demonstrated by studies documenting increased PSD-95-immunopositive puncta in both upper (39.5%) and deep layers (47.9%) of the medial prefrontal cortex in models with elevated CBLN2 expression . To establish causality, combine antibody detection with gain-of-function and loss-of-function approaches, such as comparing synaptic patterns in wildtype versus CBLN2 knockout models or in systems with regulated CBLN2 overexpression . Finally, proximity ligation assays using CBLN2 antibodies can identify direct protein-protein interactions at synapses, potentially revealing CBLN2's binding partners in the synaptogenic pathway.
Integrating CBLN2 antibody techniques with genetic models offers powerful insights into protein function. Researchers have successfully employed several complementary approaches in this domain. Transgenic models with modified CBLN2 loci (such as knockout or humanized enhancer models) provide platforms for antibody-based phenotypic analysis . For example, studies have deleted exons 1-3 of the Cbln2 gene in mice and used anti-CBLN2 immunostaining to confirm protein loss and examine consequent structural changes . Conditional knockout systems driven by tissue-specific promoters allow spatial and temporal control of CBLN2 expression, which can be monitored through antibody detection to correlate with phenotypic outcomes . CRISPR-Cas9 genome editing offers precise modification of CBLN2 regulatory elements or coding sequences, and subsequent antibody-based quantification can measure resulting expression changes. For in vivo functional analysis, AAV-mediated CBLN2 overexpression or knockdown followed by antibody staining can map region-specific functions. When analyzing these models, standardized immunohistochemical protocols are essential for reliable comparisons between genotypes. Computational analysis of antibody-labeled sections, including automated synapse counting and morphological analysis, enhances quantitative rigor when comparing genetic variants. Finally, single-cell approaches combining genetic lineage tracing with CBLN2 immunodetection can reveal cell-autonomous versus non-cell-autonomous functions in neural circuits.
CBLN2 antibodies provide valuable tools for investigating cancer mechanisms, particularly in ovarian cancer research where CBLN2 has shown significant relevance. Western blot analysis using anti-CBLN2 antibodies has revealed differential CBLN2 expression between cisplatin-resistant and cisplatin-sensitive ovarian cancer cell lines, with the chemoresistant high-grade serous carcinoma cell line A2780-CP showing 5.4-fold higher expression compared to its cisplatin-sensitive counterpart A2780-S . This suggests CBLN2 as a potential biomarker for chemoresistance. For such analyses, researchers should optimize protein extraction protocols for both cellular and secreted fractions, as CBLN2 functions as both a cytoplasmic and secreted glycoprotein . Immunohistochemistry with validated anti-CBLN2 antibodies enables evaluation of expression patterns in patient tumor samples, potentially correlating expression with clinical outcomes and treatment response. Multiplexed immunofluorescence combining CBLN2 antibodies with markers of cancer progression or chemoresistance pathways can elucidate mechanistic relationships. For translational research, developing standardized immunohistochemical scoring systems for CBLN2 expression in tumors could establish clinically relevant thresholds. Antibody-based proximity ligation assays might identify novel CBLN2 protein interactions specific to cancer contexts, while tissue microarray analysis using CBLN2 antibodies could efficiently screen expression across large patient cohorts with diverse clinical characteristics.
When investigating CBLN2 in neurodevelopmental contexts, several methodological considerations enhance research quality. Developmental timing is crucial - CBLN2 expression shows temporal specificity, requiring careful staging of samples from embryonic through postnatal periods . Region-specific analysis is essential, as CBLN2 exhibits differential expression across brain regions, with functional studies showing distinct effects in prefrontal cortex compared to somatosensory or visual areas . When using antibodies for developmental studies, verify that the specific epitope is conserved across developmental stages and not masked by stage-specific post-translational modifications. Co-immunostaining with layer-specific cortical markers helps precisely localize CBLN2 expression within developing neural circuits, as demonstrated by studies distinguishing effects in upper versus deep cortical layers . For functional analysis, combine immunohistochemistry with quantitative assessments of synaptic structures - researchers have successfully correlated CBLN2 expression with postsynaptic density using PSD-95/DLG4 and gephyrin immunostaining to measure excitatory and inhibitory synapse formation, respectively . When comparing developmental trajectories between species (especially in evolutionary studies), ensure antibodies recognize conserved epitopes to avoid species-specific detection biases. Finally, consider using clearing techniques (CLARITY, iDISCO) combined with CBLN2 immunolabeling for three-dimensional analysis of developmental expression patterns across intact brain regions.
Subcellular localization studies of CBLN2 require tailored approaches that account for the protein's diverse compartmentalization. For high-resolution mapping, combine CBLN2 antibodies with organelle-specific markers through multi-channel confocal microscopy . Previous research has successfully co-localized CBLN2 with cathepsin D-positive lysosomal structures, quantifying the degree of co-localization through confocal reconstruction across multiple tissue sections . When investigating CBLN2's dual roles as intracellular and secreted protein, employ differential permeabilization protocols to distinguish membrane-associated versus intracellular pools. For trafficking studies, pulse-chase experiments with protein synthesis inhibitors, followed by immunostaining at various time points, can track CBLN2 movement through cellular compartments. Super-resolution microscopy techniques (STED, STORM, PALM) provide nanoscale resolution of CBLN2 localization at specialized subcellular structures like synapses. For in vivo contexts, consider tissue-specific fixation optimization, as standard paraformaldehyde fixation may not preserve all CBLN2 pools equally. Electron microscopy with immunogold labeling offers ultrastructural localization but requires specialized antibodies compatible with EM processing. When analyzing co-localization data, employ quantitative metrics beyond visual assessment, such as Pearson's correlation coefficient or Manders' overlap coefficient, to ensure reproducible results across experimental conditions and biological replicates.
When developing novel applications for CBLN2 antibodies, comprehensive validation ensures reliable data. Begin with epitope mapping to confirm the antibody's binding site on CBLN2, particularly important given the protein's multiple domains and potential processing variants . For novel species applications, perform sequence alignment analysis of the target epitope across species to predict cross-reactivity before experimental validation. When adapting antibodies to new techniques (e.g., from Western blot to flow cytometry), optimize each protocol component including antibody concentration, incubation time, and buffer composition. Specificity confirmation should employ multiple approaches: knockout/knockdown controls, peptide competition assays, and comparison with alternative antibodies targeting different CBLN2 epitopes . For novel tissue applications, compare staining patterns with in situ hybridization of CBLN2 mRNA to verify correspondence between protein and transcript distribution. When establishing quantitative applications, generate standard curves using recombinant CBLN2 protein to determine detection limits and linear range. Inter-laboratory validation strengthens confidence - consider coordinated testing across multiple research teams using standardized protocols and sample sets. Finally, publish detailed validation data including positive and negative controls, antibody dilution series, and specification of exact epitope sequences to facilitate reproducibility across the research community.
Sample preparation significantly impacts CBLN2 antibody performance across applications. For tissue fixation, paraformaldehyde concentration and duration must be optimized to preserve epitope accessibility while maintaining structural integrity . Antigen retrieval methods require careful selection - heat-mediated antigen retrieval has proven effective for CBLN2 detection in paraffin sections, but optimal buffer composition (citrate vs. EDTA-based) may vary by tissue type and antibody . For protein extraction, different lysis buffers yield varying CBLN2 recovery efficiency; consider that CBLN2 exists in both soluble and membrane-associated forms, potentially requiring sequential extraction protocols . When analyzing secreted CBLN2, standardize serum-free conditioning time for consistent results across experimental groups, as demonstrated in protocols used for iTRAQ differential expression analysis . For Western blotting, sample denaturation conditions impact detection - CBLN2 forms homo- and heterodimers that may require specific reducing agent concentrations for complete dissociation . Storage conditions of both samples and antibodies influence reproducibility; establish stability profiles through time-course experiments with consistent positive controls. For challenging tissues or applications with high background, consider tissue-specific blocking reagents or signal amplification systems. Finally, batch processing of samples across experimental groups minimizes technical variability when comparing CBLN2 expression between conditions.
Reliable quantification of CBLN2 expression changes requires rigorous methodological approaches. Western blot densitometry offers a standard approach but demands careful normalization to appropriate loading controls and inclusion of calibration standards for inter-blot comparisons . For precise protein quantification, ELISA or quantitative mass spectrometry-based approaches like iTRAQ provide superior dynamic range and reproducibility, as demonstrated in secretome analysis of cancer cell lines . When analyzing tissue sections, stereological counting principles should be applied to immunohistochemical data, with systematic random sampling across regions of interest. For fluorescence-based quantification, establish standardized image acquisition parameters (exposure time, gain, offset) and include fluorescence intensity calibration standards in each imaging session. When comparing punctate structures like synapses, automated image analysis algorithms can reduce observer bias in counting PSD-95 or gephyrin-positive puncta associated with CBLN2 expression changes . Single-cell approaches such as flow cytometry or imaging flow cytometry with CBLN2 antibodies enable population-level analysis while capturing cell-to-cell variability. For longitudinal studies, consider stable isotope labeling approaches combined with CBLN2 immunoprecipitation to measure protein turnover rates. Statistical analysis should account for nested data structures (multiple measurements per subject) and include power calculations to ensure adequate sample sizes for detecting biologically relevant changes.
Integrating CBLN2 antibody data with multi-omics approaches provides comprehensive biological context. Researchers can correlate protein-level measurements from immunoassays with transcriptomic data to identify post-transcriptional regulation mechanisms affecting CBLN2 expression . Phosphoproteomics combined with CBLN2 immunoprecipitation can reveal signaling networks regulating CBLN2 function or modified by CBLN2 activity. ChIP-seq using antibodies against transcription factors identified through bioinformatic analysis (such as those binding the CBLN2 enhancer regions) helps map the regulatory network controlling CBLN2 expression . For pathway analysis, correlate CBLN2 protein levels with metabolomic profiles to identify associated metabolic shifts, particularly relevant in cancer contexts where CBLN2 shows altered expression in chemoresistant states . Spatial transcriptomics combined with CBLN2 immunohistochemistry on adjacent sections provides regional context to expression patterns across complex tissues. Network analysis integrating protein-protein interaction data with CBLN2 co-expression patterns can predict functional relationships and potential therapeutic targets. For translational research, correlate CBLN2 immunohistochemistry scores with patient genomic data to identify genetic variants associated with altered protein expression. Finally, machine learning approaches can integrate these multi-modal datasets to build predictive models of CBLN2 function and regulation, potentially identifying novel therapeutic approaches for CBLN2-associated pathologies.
Emerging technologies promise to revolutionize CBLN2 antibody-based research. Single-cell proteomics techniques will enable analysis of CBLN2 expression heterogeneity within specific cell populations, moving beyond tissue-level averages. Spatially resolved proteomics combining CBLN2 antibodies with technologies like Imaging Mass Cytometry or GeoMx Digital Spatial Profiling will map expression in complex tissue architectures while preserving spatial relationships. CRISPR-based tagging of endogenous CBLN2 with fluorescent proteins or epitope tags will facilitate live-cell imaging of CBLN2 dynamics without antibody limitations. Antibody engineering approaches, including the development of recombinant nanobodies against CBLN2, may provide superior tissue penetration and reduced background for imaging applications . Organoid models combined with CBLN2 immunostaining will enable studies of protein function in three-dimensional tissue contexts that better recapitulate in vivo development. Advances in multiplex immunohistochemistry will allow simultaneous visualization of CBLN2 alongside dozens of other markers in single tissue sections, revealing complex cellular networks. High-throughput automated immunoassay platforms will facilitate screening of CBLN2 expression across large sample cohorts, particularly valuable for biomarker development in cancer contexts . Finally, computational approaches integrating antibody-based imaging with artificial intelligence will enable automated quantification of subtle phenotypes associated with CBLN2 perturbation, potentially revealing previously unrecognized functions.