The closest matches in antibody research include:
A. LRP6 Antibodies
Low-density lipoprotein receptor-related protein 6 (LRP6) antibodies are extensively documented as critical tools for studying Wnt/β-catenin signaling pathways. Key findings include:
B. RPL6 Antibodies
Ribosomal protein L6 (RPL6) antibodies target a 33 kDa protein involved in ribosomal function and cancer biology:
Relevant antibody classes with structural or functional parallels include:
A. Anti-TMPRSS6 Antibodies
Though not directly related, TMPRSS6-targeting antibodies (e.g., MWTX-003) modulate iron homeostasis:
B. Anti-GRP78 Antibodies
PAT-SM6 (human IgM):
Key quality parameters absent for "RDL6" but critical for related antibodies:
Confirm target nomenclature (potential typographical errors for LRP6, RPL6, or TMPRSS6).
Explore antibody databases (CiteAb, AntibodyRegistry) for unpublished "RDL6" references.
Validate cross-reactivity if targeting novel epitopes in lipoprotein or ribosomal pathways.
KEGG: ath:AT1G58848
UniGene: At.22095
LRP-6 (Low-density lipoprotein receptor-related protein 6) functions as a critical membrane coreceptor for Wnt proteins, playing an essential role in the canonical Wnt/β-catenin signaling pathway that regulates numerous developmental processes and is frequently dysregulated in cancer. The importance of LRP-6 stems from its position as a key upstream mediator of Wnt signal transduction, making it an attractive target for both mechanistic studies and therapeutic intervention. When Wnt proteins bind to LRP-6 and Frizzled receptors, they trigger a signaling cascade that ultimately leads to β-catenin stabilization and nuclear translocation, activating transcription of target genes involved in cell proliferation and differentiation. Disregulated Wnt/β-catenin signaling has been directly linked to various human diseases, including different types of cancer, positioning LRP-6 as a significant research focus . Understanding LRP-6 biology through antibody-based approaches has provided crucial insights into both normal development and pathological conditions where Wnt signaling becomes aberrant.
LRP-6 antibodies can be classified into distinct functional categories based on their effects on Wnt signaling and their binding epitopes. Research has identified two major classes of LRP-6 antagonistic antibodies: one class specifically inhibits Wnt proteins represented by Wnt1, while the second class specifically inhibits Wnt proteins represented by Wnt3a . Epitope-mapping experiments have revealed that Wnt1 class-specific antibodies bind to the first propeller domain of LRP-6, whereas Wnt3a class-specific antibodies interact with the third propeller domain . This specificity corresponds with structural evidence suggesting that different Wnt family members engage distinct regions of the LRP-6 receptor. Beyond antagonistic antibodies, some LRP-6 antibodies can actually enhance Wnt signaling, functioning as agonists rather than inhibitors . The identification of these functionally diverse antibodies has significantly advanced our understanding of LRP-6 structure-function relationships and the molecular mechanisms underlying Wnt-induced LRP-6 activation, providing powerful tools for manipulating Wnt signaling experimentally.
LRP-6 antibodies serve multiple experimental purposes across various research platforms, with applications extending from in vitro cellular studies to in vivo animal models. In flow cytometry applications, LRP-6 antibodies like Mouse Anti-Human LRP-6 Monoclonal Antibody (MAB1505) can effectively detect LRP-6 expression in human cancer cell lines such as MDA-MB-231 breast cancer cells, providing quantitative measurement of receptor levels at the single-cell level . For immunocytochemistry and fluorescent imaging, these antibodies can visualize LRP-6 localization, as demonstrated in DU145 human prostate carcinoma cells where LRP-6 was detected primarily at cell surfaces and in the cytoplasm . In direct ELISA assays, human LRP-6 antibodies show high specificity with minimal cross-reactivity to related proteins, making them valuable for quantitative protein detection . Perhaps most significantly, LRP-6 antibodies have demonstrated utility in in vivo cancer models, where Wnt1 or Wnt3a class-specific anti-LRP6 antibodies specifically block growth of MMTV-Wnt1 or MMTV-Wnt3 xenografts, highlighting their potential as both research tools and therapeutic agents . Each application requires specific optimization of antibody concentration and experimental conditions to achieve reliable and reproducible results.
Validating LRP-6 antibody specificity requires a multi-faceted approach incorporating both positive and negative controls across different experimental platforms. Researchers should first assess cross-reactivity profiles through direct ELISA against a panel of related proteins, particularly focusing on LRP-5, which shares significant structural homology with LRP-6. For example, the human LRP-6 Antibody MAB1505 shows approximately 5% cross-reactivity with recombinant mouse LRP-6 but no detectable cross-reactivity with other related proteins in direct ELISAs . Cell-based validation should employ positive control cell lines with confirmed LRP-6 expression (such as MDA-MB-231 or DU145) alongside negative control cell lines where LRP-6 expression is absent or has been knocked out through CRISPR-Cas9 or similar gene editing techniques. Flow cytometry represents a particularly valuable validation approach, where researchers should compare staining with the LRP-6 antibody against appropriate isotype control antibodies to distinguish specific from non-specific binding . For immunocytochemistry applications, validation should include competitive binding assays with recombinant LRP-6 protein and assessment of staining patterns to confirm expected cellular localization patterns, such as membrane and cytoplasmic distribution observed in DU145 cells . Additional validation through Western blotting with recombinant LRP-6 protein standards and cell lysates from LRP-6 knockout lines provides further confirmation of antibody specificity and reliability.
Effective application of LRP-6 antibodies in Wnt signaling research demands careful consideration of multiple technical parameters to ensure experimental validity and interpretability. When selecting an LRP-6 antibody, researchers must first determine which Wnt pathway they wish to modulate, as different antibodies target distinct propeller domains of LRP-6 and therefore affect different Wnt protein classes - antibodies binding the first propeller domain inhibit Wnt1-class proteins while those targeting the third propeller domain inhibit Wnt3a-class proteins . Researchers should be aware of potential agonistic effects, as bivalent LRP-6 antibodies can unexpectedly sensitize cells to non-blocked classes of Wnt proteins, potentially confounding experimental interpretations . To circumvent this limitation, biparatopic LRP-6 antibodies that block both Wnt1- and Wnt3a-mediated signaling without showing agonistic activity may be preferable for comprehensive pathway inhibition . Concentration optimization is critical, as antibody effects may be dose-dependent, requiring careful titration experiments with appropriate readouts of Wnt pathway activity, such as β-catenin nuclear translocation or TCF/LEF reporter assays. When conducting in vivo studies with LRP-6 antibodies, researchers must carefully consider antibody pharmacokinetics, biodistribution, and potential immunogenicity, particularly for prolonged treatment studies in xenograft models where antibody efficacy has been demonstrated for Wnt-driven tumors .
LRP-6 antibodies exert differential effects on Wnt-dependent cellular processes due to the complex, context-dependent nature of Wnt signaling and the structural specificity of antibody-receptor interactions. The impact of LRP-6 antibodies varies substantially depending on which specific Wnt ligand-receptor interaction they disrupt, as evidenced by the finding that Wnt1 class-specific antibodies and Wnt3a class-specific antibodies bind to different propeller domains of LRP-6 and therefore selectively inhibit distinct subsets of Wnt proteins . This selectivity allows researchers to experimentally dissect the contributions of different Wnt ligands to cellular phenotypes including proliferation, differentiation, and survival. In cancer cells, these differential effects manifest as varying degrees of growth inhibition, with LRP-6 antibodies showing particular efficacy against tumors driven by the specific Wnt class they target, as demonstrated in studies where Wnt1 or Wnt3a class-specific anti-LRP6 antibodies specifically block growth of the corresponding MMTV-Wnt1 or MMTV-Wnt3 xenografts in vivo . Beyond simple growth inhibition, LRP-6 antibodies can modulate more complex phenotypes like stemness, epithelial-mesenchymal transition, and therapy resistance, which are all influenced by Wnt signaling in context-specific ways. Importantly, the agonistic potential of some LRP-6 antibodies reveals that they can sometimes enhance Wnt signaling rather than inhibit it, highlighting the complexity of receptor-antibody interactions and the need for careful experimental design when studying their effects on cellular processes .
Determining tumor susceptibility to LRP-6 antibody targeting requires a systematic analytical approach that assesses both Wnt dependency and the specific Wnt ligands driving tumor growth. Researchers should first establish Wnt pathway activation status through analysis of downstream signaling components, including immunohistochemical assessment of β-catenin nuclear localization and quantitative PCR measurement of Wnt target gene expression (e.g., AXIN2, CCND1, MYC). Next, comprehensive profiling of the specific Wnt ligands expressed in the tumor is essential, as this dictates which class of LRP-6 antibody would be most effective - Wnt1 class-specific or Wnt3a class-specific antibodies target distinct tumor types driven by different Wnt proteins . This profiling should employ RNA sequencing to identify the predominant Wnt ligands expressed, followed by functional validation using targeted knockdown of candidate Wnt genes to confirm their growth-promoting role. Ex vivo tumor slice cultures or patient-derived xenograft models provide platforms for direct testing of different LRP-6 antibodies against patient tumor samples, allowing assessment of growth inhibition, pathway suppression, and potential resistance mechanisms. Single-cell analyses of tumor heterogeneity are particularly valuable for identifying subpopulations that might respond differently to LRP-6 antibody treatment or serve as reservoirs for resistance. Additionally, researchers should evaluate potential biomarkers of response, such as receptor expression levels or pathway mutations, that might predict sensitivity to LRP-6 antibody therapy, facilitating the development of companion diagnostics for future clinical applications .
Flow cytometric analysis of LRP-6 expression requires careful optimization of antibody concentrations, staining conditions, and appropriate controls to achieve specific and reproducible results. Researchers should begin with cell preparation, ensuring single-cell suspensions through gentle enzymatic dissociation methods that preserve surface epitopes, followed by fixation protocols compatible with the specific LRP-6 antibody being used. For human cancer cell lines like MDA-MB-231, Mouse Anti-Human LRP-6 Monoclonal Antibody (MAB1505) has been successfully employed at optimized concentrations, though each laboratory should conduct titration experiments to determine ideal antibody concentrations for their specific cell system . Critical to reliable flow cytometry is the inclusion of appropriate controls, including isotype control antibodies that match the species, isotype, and conjugation status of the anti-LRP-6 antibody (such as MAB003) to establish background staining levels and set proper gating strategies . For indirect staining approaches, secondary antibody selection is crucial - Phycoerythrin-conjugated Anti-Mouse IgG Secondary Antibody (F0102B) has been successfully used with LRP-6 primary antibodies, providing sufficient signal-to-noise ratio for clear population discrimination . Data analysis should incorporate both the percentage of LRP-6 positive cells and the mean fluorescence intensity as complementary measures of expression levels. For multi-parameter flow cytometry, careful compensation must be performed when combining LRP-6 staining with other markers, particularly when evaluating LRP-6 expression in specific cellular subpopulations or correlating it with stemness or differentiation markers in complex tissues.
Effective immunocytochemistry for LRP-6 visualization requires careful attention to fixation, permeabilization, antibody selection, and imaging parameters to accurately capture receptor localization and expression patterns. Cell preparation should begin with appropriate seeding densities on coverslips or chamber slides, allowing sufficient attachment time before proceeding to fixation with either 4% paraformaldehyde (for general applications) or methanol (which may better preserve certain LRP-6 epitopes). For DU145 human prostate carcinoma cells, immersion fixation followed by careful permeabilization has successfully preserved LRP-6 epitopes for antibody detection . Primary antibody selection and concentration are critical determinants of staining quality - Mouse Anti-Human LRP-6 Monoclonal Antibody (MAB1505) has been validated at 10 μg/mL with a 3-hour room temperature incubation, though optimization for specific cell types is recommended . Secondary antibody selection should consider both the imaging platform and multiplexing needs, with fluorophore-conjugated antibodies like NorthernLights 557-conjugated Anti-Mouse IgG Secondary Antibody (NL007) providing strong signal with minimal background in LRP-6 detection protocols . Counterstaining with nuclear dyes such as DAPI facilitates interpretation of LRP-6 localization relative to cellular compartments, helping distinguish membrane from cytoplasmic staining as observed in DU145 cells . For co-localization studies examining LRP-6 interaction with Wnt pathway components, careful selection of compatible antibody pairs from different host species is essential to avoid cross-reactivity. Image acquisition parameters should be optimized for the specific cellular distribution pattern of LRP-6, with appropriate z-stack collection to capture membrane localization, and consistent exposure settings across experimental conditions to enable quantitative comparisons of expression levels.
Epitope mapping for LRP-6 antibodies demands sophisticated methodological approaches that account for the complex structural features of this multi-domain receptor and the functional implications of antibody binding sites. Researchers should begin with computational analysis of LRP-6 structure, identifying distinct domains like the multiple propeller domains that have demonstrated functional significance in Wnt binding . For experimental epitope mapping, domain-specific recombinant protein fragments representing different regions of LRP-6 (such as individual propeller domains) provide a foundation for initial epitope localization through direct binding assays. Peptide arrays or phage display libraries expressing overlapping peptide fragments can further refine epitope identification to specific amino acid sequences within the larger domains. Functional classification of antibodies based on their effects on different Wnt proteins (e.g., Wnt1-class specific versus Wnt3a-class specific) should be correlated with binding domain information to establish structure-function relationships, as has been demonstrated for antibodies binding the first versus third propeller domains of LRP-6 . Site-directed mutagenesis of key residues within putative epitopes, followed by binding affinity measurements, provides definitive validation of specific contact points between antibody and receptor. For complex epitopes spanning multiple domains or conformational epitopes, hydrogen-deuterium exchange mass spectrometry offers powerful insights into antibody-receptor interactions without requiring crystallization. Advanced structural techniques including X-ray crystallography or cryo-electron microscopy of antibody-LRP-6 complexes provide the most detailed epitope information, revealing not only binding sites but also potential conformational changes induced by antibody binding that might explain functional effects on receptor activity and Wnt signaling outcomes .
Resistance to LRP-6 antibody therapy may emerge through multiple mechanisms, requiring multifaceted strategies to maintain therapeutic efficacy in Wnt-driven cancers. Primary resistance may occur in tumors expressing multiple Wnt ligands from different classes, as monospecific antibodies targeting either the first or third propeller domain of LRP-6 would only block signaling from a subset of these ligands . This challenge can be addressed through biparatopic antibodies that simultaneously target multiple propeller domains, providing comprehensive pathway inhibition regardless of the specific Wnt ligands driving tumor growth . Acquired resistance might develop through selection for tumor cell subpopulations that activate Wnt signaling downstream of LRP-6, such as through mutations in APC, AXIN, or β-catenin, bypassing the receptor-level blockade imposed by LRP-6 antibodies. Combination therapies targeting both upstream (LRP-6) and downstream components of the Wnt pathway represent a promising approach to prevent or overcome such resistance mechanisms. Tumor microenvironment adaptations may also contribute to resistance, as stromal cells can produce alternative Wnt ligands or other growth factors that compensate for blocked Wnt signaling pathways. Sequential or alternating therapy with different classes of LRP-6 antibodies might prevent the emergence of resistant clones that would be selected by continuous treatment with a single antibody class. Rational combinations with other targeted therapies, immunotherapies, or conventional chemotherapies should be explored based on mechanistic understanding of pathway interactions and potential synthetic lethality relationships with inhibited Wnt signaling. Patient-derived xenograft models and organoid cultures can serve as valuable platforms for testing these resistance mechanisms and therapeutic strategies in preclinical settings before advancing to clinical trials of LRP-6 antibody therapies .
Determining antibody specificity between the highly homologous LRP-6 and LRP-5 receptors requires comprehensive analytical approaches that account for structural similarities while detecting critical differences between these related proteins. Comparative direct ELISA represents a foundational approach, where purified recombinant LRP-6 and LRP-5 proteins are tested in parallel against the antibody of interest, establishing binding profiles and quantifying cross-reactivity percentages, as demonstrated for the Human LRP-6 Antibody MAB1505 which shows minimal cross-reactivity with related proteins . Surface plasmon resonance (SPR) provides more detailed kinetic and thermodynamic parameters of antibody binding to both targets, revealing not only binding magnitude but also association and dissociation rates that may differ between LRP-6 and LRP-5 interactions. Epitope mapping using domain swapping or chimeric LRP-5/LRP-6 constructs can pinpoint regions responsible for differential antibody recognition, particularly valuable when targeting the propeller domains which may have structural differences despite sequence homology. Cell-based specificity assays using cells expressing either LRP-6 or LRP-5 (or ideally single and double knockout lines with reconstituted expression) provide functional validation in a more physiologically relevant context than purified proteins alone. Competition binding studies where unlabeled LRP-5 is used to compete with labeled LRP-6 (and vice versa) for antibody binding offer quantitative measures of relative affinity and specificity. Structural biology approaches including X-ray crystallography or cryo-electron microscopy of antibody-antigen complexes provide the most definitive evidence of binding specificity by revealing atomic-level interactions that differentiate binding to LRP-6 versus LRP-5. Computational approaches utilizing homology modeling and molecular dynamics simulations can predict antibody binding to both targets based on structural information, guiding experimental design and helping interpret experimental results in structural contexts .
Correlating LRP-6 antibody epitopes with functional effects on Wnt signaling requires integrated structural and functional analyses that connect specific binding sites to downstream signaling outcomes. Researchers should begin with precise epitope mapping using techniques such as hydrogen-deuterium exchange mass spectrometry, X-ray crystallography, or mutagenesis studies to identify the exact binding regions of different antibodies on the LRP-6 receptor. Once epitopes are mapped, classification of antibodies based on which propeller domain they bind provides initial functional insights, as antibodies binding the first propeller domain typically inhibit Wnt1-class proteins while those binding the third propeller domain inhibit Wnt3a-class proteins . Functional validation requires measuring multiple Wnt signaling readouts, including β-catenin stabilization, nuclear translocation, TCF/LEF reporter activation, and target gene expression after treatment with epitope-defined antibodies. Comparison of multiple antibodies binding the same epitope but with different affinities or binding modes can reveal how specific aspects of antibody-receptor interaction, beyond simple binding location, influence signaling outcomes. Structure-function relationships can be further elucidated by examining how antibodies affect LRP-6 receptor clustering, internalization, and interaction with other pathway components like Frizzled receptors or Wnt ligands. Advanced microscopy techniques such as single-molecule tracking or FRET can visualize how antibody binding to specific epitopes alters receptor dynamics and complex formation in living cells. Computational modeling incorporating protein structures, binding energetics, and pathway dynamics can generate testable hypotheses about how specific epitope binding translates to altered receptor conformation and function. The comparison of monoclonal versus biparatopic antibodies targeting multiple epitopes simultaneously provides particularly valuable insights into how different domains of LRP-6 cooperate in signal transduction, as evidenced by the finding that biparatopic antibodies can block both Wnt1- and Wnt3a-mediated signaling without showing the agonistic effects sometimes observed with monospecific antibodies .