While not an antibody, the LC16m8 strain is a third-generation live-attenuated vaccine derived from the Lister strain of vaccinia virus. It has been studied for smallpox and mpox prevention:
CD16 (FcγRIII) is a low-affinity IgG receptor critical for antibody-dependent cellular cytotoxicity (ADCC). Commercial antibodies targeting CD16 include:
Clone TH-16 (Catalog #MA1100) is a mouse monoclonal antibody targeting tyrosine hydroxylase (TH), a rate-limiting enzyme in dopamine synthesis:
| Parameter | Details | Source |
|---|---|---|
| Host | Mouse IgG1 | |
| Applications | Western blot (59 kDa band), IHC (rat brain tissues) | |
| Cross-Reactivity | Human, mouse, rabbit, rat (no off-target binding observed). |
Epitope: Binds a 26-amino acid segment of MUC16’s proximal ectodomain via β-turns and β-hairpin motifs .
Therapeutic Potential: Demonstrated utility as an antibody-drug conjugate (ADC) and chimeric antigen receptor (CAR) .
Binding Interface: 876 Ų buried surface area with 10 salt bridges/hydrogen bonds stabilizing the interaction .
CDR Loops: 22 residues from complementarity-determining regions (CDRs) mediate contact .
Verify Terminology: Confirm whether "LCR16" refers to a novel target, a typo (e.g., LC16, CD16), or a proprietary compound not yet published.
Exploratory Assays: If investigating a new antibody, consider:
Phage Display Libraries for epitope mapping.
Surface Plasmon Resonance (SPR) for affinity measurements.
Database Searches: Query repositories like UniProt (ID: P08637 for CD16) or ClinicalTrials.gov for ongoing studies.
The LCR16 antibody refers to antibodies targeting or related to the Long Control Region (LCR) of Human Papillomavirus 16 (HPV16). The LCR is a critical regulatory region of the HPV16 genome that controls viral gene expression. Antibodies targeting this region are valuable for studying transcriptional regulation of HPV16 genes, particularly the oncogenes E6 and E7. These antibodies can be used to investigate how proteins like SOX2 interact with the HPV16-LCR and subsequently affect viral gene expression in a squamous cell carcinoma context .
Several key experimental techniques are employed to study antibody binding to the HPV16-LCR region:
Electrophoretic Mobility Shift Assays (EMSA): Used to detect protein-DNA interactions, including super-shift analysis with specific antibodies to confirm binding of factors like SOX2 to the HPV16-LCR.
Chromatin Immunoprecipitation (ChIP) Assays: Used to determine if proteins bind to specific DNA sequences in vivo. Cells are cross-linked with formaldehyde, lysed, and sonicated to fragment chromatin, followed by immunoprecipitation with specific antibodies against proteins of interest that may bind to the LCR .
Multi-Site-Directed Mutagenesis: Employed to evaluate the functional significance of specific binding sites within the HPV16-LCR .
When validating antibody specificity for HPV16-LCR research, several controls should be implemented:
Positive Binding Control: Use antibodies against proteins known to bind the LCR (e.g., anti-p65 antibodies as LCR-binding controls) .
Negative Binding Control: Include antibodies against proteins not expected to bind the LCR (e.g., anti-CTCF antibodies as non-binding controls) .
Background Control: Use normal rabbit IgG antibody to determine non-specific binding .
Cold Probe Competition: For EMSA experiments, include competition with specific (CS) and non-specific (NS) cold probes at various fold excesses (e.g., 50-fold and 250-fold) to confirm binding specificity .
Western Blot Validation: Confirm antibody specificity against the target protein using appropriate protein load controls (e.g., α-actinin, β-actin, or GAPDH) .
Framework mutations in antibodies can significantly alter their structural dynamics and binding properties, which has important implications for HPV16-LCR research:
VH-VL Interface Angle Shifts: Framework mutations can cause significant shifts in the distribution of variable heavy chain-variable light chain (VH-VL) interface angles, which affects antibody flexibility and function. These changes are not evident in static crystal structures but become apparent in solution-state dynamics .
Local Flexibility Changes: Mutations alter the antibody's solvent accessible surface area (SASA), leading to changes in local flexibility. This affects how antibodies interact with targets like the HPV16-LCR .
Conformational Ensemble Expansion: Framework mutations can increase the number of stable macrostates available in the antibody's conformational ensemble, potentially providing advantages during affinity maturation by allowing exploration of larger conformational spaces when encountering diverse antigens .
Functional Consequences: These structural dynamics influence antibody specificity, stability, and binding breadth - all critical factors in developing effective antibodies for HPV16-LCR research .
Researchers should consider these dynamic aspects when designing or selecting antibodies for HPV16-LCR investigations, moving beyond the "static dogma" of antibody structures toward a more dynamic understanding .
Advanced computational methods can significantly enhance LCR16 antibody research through:
Homology Modeling Workflows: Researchers can predict antibody structure using guided homology modeling that incorporates de novo CDR loop conformation prediction, allowing for structural modeling directly from sequence data .
Batch Modeling for Variant Analysis: Accelerated model construction for parent sequences and variants enables efficient comparison of multiple antibody candidates targeting the HPV16-LCR .
Protein-Protein Docking: Ensemble protein-protein docking can predict antibody-antigen complex structures, helping researchers understand and predict interactions between antibodies and the HPV16-LCR .
Epitope Mapping Enhancement: Computational approaches can enhance resolution of experimental epitope mapping data from peptide to residue-level detail, providing more precise information about binding sites .
Liabilities Prediction: Early identification of potential developmental issues through modeling and structure characterization, including detection of potential hotspots for aggregation and post-translational modification sites .
In Silico Engineering: Accurate prediction of how residue substitutions impact binding affinity, selectivity, and thermostability allows for rapid identification of high-quality variants using techniques like Residue Scan FEP+ with lambda dynamics .
Optimizing antibodies for detecting SOX2 binding to HPV16-LCR requires several methodological considerations:
Binding Site Identification: First identify the putative SOX2 binding sites within the HPV16-LCR. Research has identified multiple SOX2 binding sites (S1, S2, and S3) within the HPV16-LCR that are essential for SOX2-mediated repression .
Antibody Selection Criteria:
Choose antibodies with proven specificity for SOX2, such as those validated in previous studies (e.g., rabbit anti-SOX2 antibody from Santa Cruz Biotechnology, sc-20088 or Abcam, ab59776) .
Consider antibodies that maintain their recognition capabilities under the conditions required for techniques like EMSA and ChIP.
Experimental Validation Approach:
Perform EMSA with cold probe competition to confirm binding specificity to the identified SOX2 sites.
Conduct ChIP assays to verify in vivo binding using cross-linking with 1% formaldehyde and appropriate sonication to achieve optimal chromatin fragmentation (300-500 bp) .
Validate functional relevance through reporter assays measuring transcriptional repression of the HPV16-LCR by SOX2.
Functional Impact Assessment: After confirming SOX2 binding, analyze downstream effects on E6 and E7 oncogene expression to establish the functional significance of the binding in the context of HPV16 regulation .
Optimizing ChIP assays for HPV16-LCR research requires careful attention to several methodological details:
| Parameter | Recommended Conditions | Rationale |
|---|---|---|
| Cross-linking | 1% formaldehyde | Provides optimal protein-DNA fixation while maintaining epitope accessibility |
| Quenching | 0.125 M glycine | Stops cross-linking reaction and prevents over-fixation |
| Cell lysis | Buffer containing 1% SDS | Effectively disrupts cell membranes while preserving protein-DNA complexes |
| Sonication | Adjust to achieve 300-500 bp fragments | Fragment size is crucial for efficient immunoprecipitation and resolution |
| Primary antibody selection | Anti-SOX2: Abcam ab59776 (1:2000 dilution) | Previously validated for HPV16-LCR binding studies |
| Controls | Anti-p65 (positive control), Anti-CTCF (negative control), Normal rabbit IgG (background) | Essential for validating specificity and quantifying background |
| Antibody incubation | Overnight at 4°C with rotation | Allows for optimal antibody-antigen interaction |
| Wash stringency | Progressive stringency washes | Reduces non-specific binding while preserving specific interactions |
| Detection method | qPCR with LCR-specific primers | Provides quantitative measurement of enrichment |
Following these optimized conditions will maximize the sensitivity and specificity of ChIP assays for investigating protein interactions with the HPV16-LCR .
When investigating new potential regulators of the HPV16-LCR, a systematic approach to antibody validation is essential:
In silico Analysis:
Antibody Selection and Initial Validation:
Verify antibody specificity via Western blot against cell lines with known expression levels of the target protein.
Confirm recognition of the native protein by immunoprecipitation.
Validate antibody performance in fixed cells via immunofluorescence to ensure nuclear localization for transcription factors.
Binding Validation Workflow:
In vivo Binding Confirmation:
Implement ChIP assays following the optimized protocol described in section 3.1.
Include appropriate positive and negative controls for binding.
Quantify enrichment relative to input and IgG controls.
Functional Impact Assessment:
Cross-validation:
Use complementary techniques such as DNA-protein pull-down assays.
Consider advanced methods like CUT&RUN or CUT&Tag for higher resolution binding profiles.
This systematic approach ensures reliable identification and characterization of new HPV16-LCR regulators while minimizing false positives.
Non-specific binding is a common challenge in antibody-based HPV16-LCR studies. Here are methodological approaches to address this issue:
Antibody Selection Optimization:
Compare multiple commercially available antibodies targeting the same protein (e.g., SOX2).
Validate each using Western blot against positive and negative control cell lines.
Consider using monoclonal antibodies for higher specificity in certain applications.
EMSA Optimization Strategies:
Adjust binding reaction conditions (salt concentration, pH, temperature) to reduce non-specific interactions.
Implement a two-step competition approach: pre-incubate with non-specific competitor DNA before adding specific probe.
Increase poly(dI-dC) concentration to suppress non-specific DNA-protein interactions.
Optimize protein extract concentration to maintain signal-to-noise ratio .
ChIP Assay Refinement:
Implement more stringent washing steps (increase salt concentration progressively).
Add pre-clearing steps with protein A/G beads and non-specific IgG.
Consider dual cross-linking with DSG (disuccinimidyl glutarate) and formaldehyde for transcription factors.
Test sonication conditions to ensure optimal chromatin fragmentation (300-500 bp) .
Quantitative Controls Implementation:
Include parallel ChIP reactions with IgG from the same species as the primary antibody.
Measure enrichment at genomic regions not expected to bind the target protein.
Calculate signal-to-noise ratios relative to these controls for objective assessment.
Signal Validation Approaches:
Confirm binding with a second antibody targeting a different epitope of the same protein.
Use gene knockdown/knockout to demonstrate specificity (signal should be reduced/absent).
Implement peptide competition assays where specific blocking peptides abolish true binding signals.
Following these methodological refinements will help distinguish specific from non-specific signals in antibody-based HPV16-LCR studies.
When faced with contradictory results from different antibodies targeting the same protein in HPV16-LCR studies, researchers should implement a systematic resolution approach:
Comparative Antibody Profiling:
Document epitope locations for each antibody, as differences may explain discrepancies.
Verify each antibody's validation data, preferring those with knockout/knockdown validation.
Compare performance across multiple techniques (Western blot, IP, ChIP, IF) as some antibodies work better in specific applications .
Orthogonal Validation Strategies:
Implement complementary non-antibody techniques (e.g., DNA affinity precipitation or CRISPR-based approaches).
Use tagged protein expression systems where possible to detect binding with anti-tag antibodies.
Apply techniques like CUT&RUN or CUT&Tag that offer higher resolution and sensitivity.
Binding Site Microenvironment Analysis:
Consider chromatin accessibility at the putative binding site using ATAC-seq data.
Evaluate potential post-translational modifications that might affect antibody recognition.
Assess potential protein-protein interactions that might mask epitopes in specific contexts.
Physiological Context Examination:
Test binding under different cellular conditions (e.g., differentiation states of keratinocytes).
Evaluate binding in multiple HPV-positive cell lines with different viral integration statuses.
Consider the impact of cell cycle on binding patterns.
Quantitative Comparison Framework:
Develop a scoring system comparing antibodies across multiple parameters (specificity, sensitivity, reproducibility).
Generate concentration-dependent binding curves to assess relative affinities.
Use statistical methods to evaluate the significance of observed differences.
Consensus-Building Approach:
Consider all results collectively, giving more weight to findings confirmed by multiple methods.
Report all data transparently, including contradictory results, in publications.
Discuss limitations and potential explanations for discrepancies.
Monoclonal antibody technologies offer significant potential for advancing HPV16-LCR binding research through several innovative approaches:
Novel Monoclonal Development Strategies:
Humanized immune system mouse models can generate human-compatible antibodies directly, eliminating the need for re-engineering antibodies for human applications .
Immunization with multiple different elements of the HPV16-LCR allows the mouse immune system to naturally develop diverse antibodies targeting different aspects of the regulatory region .
Enhanced Specificity and Characterization:
Monoclonal antibodies offer superior specificity compared to polyclonal alternatives, reducing background and cross-reactivity in techniques like ChIP and EMSA.
The ability to isolate and characterize hundreds of different antibodies (comparable to the 300 antibodies isolated in antimicrobial resistance research) enables identification of those with optimal recognition properties for specific HPV16-LCR binding proteins .
Temporal Evolution Studies:
Monoclonal antibodies can be developed against HPV16-LCR isolates from different timepoints, enabling studies of how the LCR evolves over time in response to selective pressures.
This approach parallels the antimicrobial resistance research where antibodies remained effective against bacteria isolated ten years apart, suggesting applications for studying HPV16 evolution .
Therapeutic Development Potential:
Beyond research applications, monoclonal antibodies targeting key regulators of HPV16-LCR activity could potentially be developed into therapeutic agents.
The established safety profile of monoclonal antibodies and existing production technologies suggest this could become a viable approach within a few years, similar to the timeline projected for antimicrobial applications .
Multi-faceted Target Recognition:
Cocktails of monoclonal antibodies could be developed to simultaneously target multiple key regulators of HPV16-LCR, providing more comprehensive pathway inhibition.
This strategy could address potential resistance mechanisms that might arise from targeting single factors.
The Patent and Literature Antibody Database (PLAbDab) offers powerful resources that can enhance HPV16-LCR research through:
Extensive Sequence Resource Utilization:
Access to 150,000 paired antibody sequences from over 10,000 small-scale studies provides unprecedented opportunities to identify antibodies with desired properties for HPV16-LCR research .
Researchers can leverage this database, which is at least an order of magnitude larger than other non-NGS databases of paired antibody sequences .
Multi-modal Search Capabilities:
Sequence identity searches using KA-search allow researchers to identify antibodies with structural similarities to those with known effectiveness in HPV16-LCR research .
Structural similarity searches enable identification of functionally similar antibodies even when sequence identity is lower .
Keyword searching facilitates finding antibodies from studies targeting similar viral regulatory elements .
Source Attribution and Additional Information:
Direct links to source materials for each sequence enable researchers to obtain additional context and experimental details about antibodies of interest .
This feature is particularly valuable for understanding the original application and validation methods used for potentially relevant antibodies .
Specialized Research Application:
The database's statistical information about antibody distribution by source (approximately 75% from patents, 20% from scientific literature) helps researchers understand the landscape of available antibodies .
Historical trends showing 10,000-30,000 new antibody sequences published annually over the past 5 years indicate a growing resource for HPV16-LCR research .
Practical Implementation Approach:
Researchers can implement a methodical workflow starting with identifying antibodies used in similar viral studies through keyword searches.
Following identification, sequences can be analyzed for CDR structures most likely to recognize HPV16-LCR binding proteins.
Selected candidates can then be synthesized or acquired for experimental validation in HPV16-LCR binding studies.
| Search Method | Search Criteria for HPV16-LCR Research | Expected Value |
|---|---|---|
| Keyword | "HPV" OR "papillomavirus" OR "transcription factor binding" | Identify studies with relevant biological context |
| VH identity | Sequence homology to validated antibodies (e.g., SOX2 antibodies) | 15-25% relevant hits based on database statistics |
| VH+VL identity | Combined heavy and light chain matching to known effective antibodies | 15-18% highly relevant candidates |
| CDR structure | Recognition patterns similar to transcription factor binding antibodies | 40-50% potentially useful structural analogs |
This systematic approach to database utilization can significantly accelerate antibody discovery for HPV16-LCR research while reducing development time and costs .