IL-16 functions as a chemoattractant cytokine and modulator of T-cell activation, proposed as a ligand for the CD4 co-receptor. It plays a crucial role in mediating T cell activation, chemotaxis, and proliferation. The active form of IL-16 is detected at sites of TH1-mediated inflammation, such as those observed in autoimmune diseases, ischemic reperfusion injury (IRI), and tissue transplant rejection. Structurally, IL-16 contains a characteristic PDZ domain with a defined globular structure and peptide-binding site between the αB and βB structural elements. Unlike typical PDZ domains, IL-16's structure reveals a tryptophan residue that obscures the recognition groove, which has important implications for its function and antibody targeting .
Anti-IL-16 antibodies function by binding to IL-16 and inhibiting its interaction with target receptors. The 14.1 monoclonal antibody binds to IL-16 through a mechanism that requires a conformational change in the IL-16 PDZ domain. This involves rotation of the αB-helix, movement of the peptide groove-obscuring tryptophan residue, and opening of the binding site for interaction. Surprisingly, the antibody binding site does not directly overlap with residues involved in CD4 binding (Arg 616, Arg 617, and Lys 618), suggesting that its inhibitory activity occurs through an allosteric mechanism or by preventing proper IL-16 presentation to CD4. When the antibody binds, it induces structural changes that likely disrupt IL-16's ability to recruit and activate CD4+ cells, thereby reducing TH1-type inflammatory responses .
IL-16 dysregulation is associated with numerous inflammatory and autoimmune conditions. Elevated IL-16 expression levels have been documented in serum from patients with rheumatoid arthritis, multiple sclerosis, and systemic lupus erythematosus. These conditions share common pathological features of chronic inflammation and autoimmune reactivity. Additionally, IL-16 plays important roles in inflammatory processes such as ischemic reperfusion injury and tissue transplant rejection. The involvement of IL-16 in T cell-mediated inflammation makes it a relevant target in various inflammatory diseases. Research also indicates associations between IL-16 and the development of several cancers, further expanding the therapeutic potential of IL-16-targeting strategies .
IL-16 contains a characteristic PDZ domain with a defined globular structure. This domain includes a peptide-binding site located in a groove between the αB and βB structural elements and a highly conserved carboxylate-binding loop. A distinctive feature of IL-16's PDZ domain is the presence of a tryptophan residue (Trp600) that obscures the recognition groove, unlike other PDZ domains. The structure of the 14.1Fab fragment in complex with IL-16 reveals that antibody binding requires a conformational change in this domain, involving rotation of the αB-helix and movement of the tryptophan residue to open up the binding site. The binding interface buries 876 Ų of antibody protein surface and 853 Ų of IL-16 surface, which is typical for antibody-target interactions. The interface features a mixture of polar and hydrophobic contacts, including eight aromatic side chains from the CDR loops .
Researchers can employ anti-IL-16 antibodies as powerful tools to dissect inflammatory pathways by selectively neutralizing IL-16 activity in experimental systems. When designing such experiments, consider using the well-characterized clone 14.1 antibody that cross-reacts with both human and murine IL-16, allowing for translational research across species. To investigate IL-16's role in specific pathways, researchers should establish baseline IL-16 expression in their model systems before antibody treatment, utilizing ELISA with paired antibodies (such as clone 17.1 for coating and biotinylated clone 14.1 for detection). For in vitro studies, T cell chemotaxis assays represent a functional readout for antibody efficacy, measuring migration inhibition when cells are pre-incubated with neutralizing antibodies. In more complex in vivo inflammatory models, administer the antibody systematically or locally to tissues of interest, then assess multiple parameters including immune cell infiltration, cytokine production, and clinical manifestations of disease. Flow cytometry analysis using anti-IL-16 antibodies alongside markers for T cell activation can reveal how IL-16 blockade affects downstream signaling cascades .
When designing experiments to evaluate IL-16 antibody efficacy in disease models, researchers must carefully consider several critical factors. First, select appropriate disease models that feature known IL-16 involvement, such as models of ischemic reperfusion injury, autoimmune conditions like experimental autoimmune encephalomyelitis (EEA), or T cell-mediated renal injury. Establish clear endpoints that specifically measure IL-16-dependent pathology rather than general inflammation. Include proper controls, including isotype-matched antibodies to rule out non-specific effects. Determine optimal antibody dosing, timing, and administration route through pilot studies—antibody administration may be required before disease induction for preventative studies or after disease establishment for therapeutic assessment. Monitor antibody pharmacokinetics and tissue distribution to ensure target engagement. Measure both IL-16 levels and activity using appropriate biomarkers, such as T cell chemotaxis or CD4+ cell activation status. Finally, complement antibody studies with genetic approaches (e.g., IL-16 knockout or knockdown models) to validate antibody-specific findings and rule out off-target effects .
The structural changes in IL-16 induced by antibody binding profoundly affect its molecular interactions through several mechanisms. When the 14.1 antibody binds to IL-16, it triggers a significant conformational change in the PDZ domain, which involves rotation of the αB-helix and movement of the Trp600 residue that normally obscures the peptide-binding groove. This structural rearrangement exposes a cryptic peptide binding site that may be involved in interactions with as-yet-unidentified molecular partners. Although the antibody binding site does not directly overlap with the CD4 binding region, the conformational changes may allosterically alter the presentation of CD4 binding residues (Arg616, Arg617, and Lys618). The importance of the Trp600 residue was demonstrated through mutation studies showing that the W600A variant exhibited a 10-fold decrease in antibody binding affinity, indicating that the network of Van der Waals interactions between the antibody CDR-H3 loop and the hydrophobic pocket formed by IL-16 residues (including Trp600) is energetically favorable despite the conformational change penalty. These structural insights suggest that designing therapeutics targeting this conformational plasticity could offer new approaches for modulating IL-16 activity .
Recent developments in engineering recombinant anti-IL-16 antibodies have significantly advanced their research applications. Researchers have successfully created chimeric antibodies, such as the anti-hIL-16-hIgG1, which combines the original mouse-derived variable regions of the 14.1 clone with a human IgG1 constant region, resulting in approximately 65% human sequence content. These recombinant antibodies are now produced in Chinese hamster ovary (CHO) cells rather than hybridoma systems, ensuring reliability and lot-to-lot reproducibility while preventing common hybridoma-related drawbacks like the expression of non-relevant mAbs with aberrant light chains. This production method in animal-free facilities with defined media has yielded antibodies with improved characteristics, including low aggregation rates (<5%), which enhances their stability and functional consistency in research applications. For specialized research needs, biotinylated versions of anti-IL-16 antibodies have been developed for detection in sandwich ELISA systems, with established optimal concentrations for coating (1 μg/mL) and detection (0.125-0.25 μg/mL). The continued refinement of these recombinant technologies provides researchers with increasingly reliable and versatile tools for studying IL-16 biology .
The optimal protocols for using anti-IL-16 antibodies vary by experimental application, with each requiring specific considerations for maximum effectiveness. For ELISA applications, researchers should employ a sandwich ELISA format using clone 17.1 (M160E) as the coating antibody at 1 μg/mL and biotinylated clone 14.1 (M161B) as the detection antibody at 0.125-0.25 μg/mL. This combination has been validated with recombinant IL-16 protein standards. For Western blotting, use the M160E antibody which detects IL-16 at its predicted molecular weight of approximately 142 kDa, with overnight primary antibody incubation at 4°C for optimal results. In flow cytometry experiments analyzing IL-16 interaction with CD4, incubate CD4+ve cells with IL-16 (concentration range: 3.3 ng/ml to 33 mg/ml) for 2 hours at 4°C, followed by a secondary detection system. For neutralization assays, pre-incubate IL-16 with the 14.1 antibody before adding to cell cultures to assess inhibition of chemotaxis or other IL-16-dependent functions. In immunoprecipitation studies, use approximately 5 μg of antibody per 500 μg of total protein, allowing overnight binding at 4°C before adding protein A/G beads. For each application, researchers should always include appropriate isotype controls and validate antibody specificity using IL-16 knockout or depleted samples .
To optimize IL-16 antibody concentration and experimental conditions across different assay systems, researchers should follow a systematic approach tailored to each application. For ELISA, perform antibody titration experiments with a standard curve of recombinant IL-16 protein to determine the optimal coating antibody concentration (typically starting at 1-5 μg/mL) and detection antibody concentration (usually 0.125-1 μg/mL). Test different blocking agents (BSA, non-fat milk, commercial blockers) to minimize background while maintaining sensitivity. For neutralization assays, establish a dose-response curve using serial dilutions of the antibody (starting from 10 μg/mL) against a fixed concentration of IL-16, and identify the minimum concentration achieving >90% inhibition. When optimizing flow cytometry conditions, titrate antibody concentrations to identify the concentration yielding maximum signal-to-noise ratio, typically testing ranges from 0.1-10 μg/mL. For Western blotting, optimize primary antibody concentration (usually 0.1-5 μg/mL), incubation time (1 hour at room temperature or overnight at 4°C), and washing conditions. For each new experimental system or biological sample type, perform cross-reactivity tests and validate antibody specificity using known positive and negative controls. Finally, determine optimal storage conditions for working antibody solutions (typically at 4°C with preservatives for short-term or aliquoted and frozen for long-term storage) to maintain consistent performance across experiments .
Multiple complementary approaches should be employed to rigorously validate anti-IL-16 antibody specificity and functionality. Begin with Western blot analysis to confirm that the antibody detects a protein of the expected molecular weight (approximately 142 kDa for IL-16) in samples with known IL-16 expression. Perform immunoprecipitation followed by mass spectrometry to verify that the antibody captures the intended target. Include appropriate controls such as IL-16 knockout/knockdown samples or recombinant IL-16 for positive control. Test cross-reactivity across species when working with antibodies like clone 14.1 that react with both human and mouse IL-16. Validate functional neutralization ability through T cell chemotaxis assays, where pre-incubation of IL-16 with the antibody should inhibit migration. Conduct competition assays with unlabeled and labeled antibodies to confirm binding to the same epitope. Perform epitope mapping through techniques like hydrogen-deuterium exchange mass spectrometry or structural analysis to confirm binding to the expected region of IL-16. Compare results across multiple anti-IL-16 antibody clones (such as 14.1 and 17.1) targeting different epitopes to strengthen confidence in observations. Finally, correlate antibody binding with known biological effects, such as reduction in TH1-type inflammatory responses in relevant disease models .
Researchers can effectively combine IL-16 antibodies with other research tools through a multi-modal approach for comprehensive pathway analysis. Pair neutralizing IL-16 antibodies with specific inhibitors of downstream signaling molecules to determine the hierarchical relationship between IL-16 and other pathway components. Combine antibody treatments with transcriptomic or proteomic profiling to identify global changes in gene or protein expression following IL-16 neutralization, revealing both direct and indirect targets. Use IL-16 antibodies alongside CRISPR-Cas9 gene editing to create cellular models with modified IL-16 signaling components, allowing precise dissection of the pathway. Implement multiplex cytokine assays to measure how IL-16 blockade affects the broader inflammatory microenvironment. Develop reporter cell lines expressing fluorescent or luminescent markers under the control of IL-16-responsive elements, providing real-time readouts when combined with antibody treatments. For in vivo studies, combine antibody administration with intravital microscopy to visualize dynamic cellular responses to IL-16 neutralization. Integrate computational modeling with experimental antibody data to predict pathway responses under different conditions. When studying IL-16 involvement in T cell function, combine antibody treatments with flow cytometric analysis of multiple activation markers to create a comprehensive profile of immune cell responses. This integrated approach generates a more complete understanding of IL-16 biology than could be achieved with antibodies alone .
When faced with contradictory results between different anti-IL-16 antibody clones, researchers should systematically investigate several potential explanations. First, recognize that different clones (such as 14.1 and 17.1) target distinct epitopes on IL-16, which may be differentially accessible depending on the experimental conditions or the conformational state of IL-16. The structure of the 14.1Fab-IL-16 complex reveals that this antibody induces significant conformational changes in IL-16, which may not occur with other antibodies. Compare the experimental methods in detail, as antibody performance can vary dramatically between applications (ELISA, Western blot, neutralization assays). Examine antibody format differences—whether you're using full IgG, Fab fragments, or recombinant versions with human constant regions versus original mouse antibodies. Verify antibody quality with positive and negative controls to rule out degradation or batch variation issues. Consider the biological context, as IL-16 may interact with different binding partners in different cell types or disease states, potentially masking certain epitopes. Test for potential cross-reactivity with other proteins, especially other PDZ domain-containing molecules. Finally, resolve contradictions by employing orthogonal methods that don't rely on antibodies, such as genetic approaches, to validate or refute antibody-based findings .
Several common pitfalls can compromise IL-16 antibody experiments, but researchers can implement specific strategies to avoid them. One major issue is improper antibody storage, which leads to degradation and inconsistent results—prevent this by storing antibodies according to manufacturer recommendations, typically aliquoted at -20°C to avoid freeze-thaw cycles. Non-specific binding can generate false positives; combat this by optimizing blocking conditions and including appropriate isotype controls in all experiments. Neglecting the importance of epitope accessibility in different applications can lead to contradictory results—the unique conformational change required for 14.1 antibody binding illustrates how sample preparation can affect epitope exposure. Inadequate validation of antibody specificity remains problematic; always confirm specificity using multiple methods, including Western blotting with positive and negative controls. Overlooking the critical Trp600 residue in IL-16 structure when interpreting binding data is a specific technical pitfall, as this residue significantly impacts antibody binding affinity. Cross-reactivity between species must be explicitly tested rather than assumed, even though the 14.1 clone has documented cross-reactivity with mouse and human IL-16. Finally, failure to account for endogenous IL-16 in complex biological samples can confound neutralization experiments—quantify baseline IL-16 levels before adding recombinant protein or antibody to accurately interpret results .
Differentiating between specific and non-specific effects of IL-16 antibodies in complex biological systems requires a multi-faceted approach. Always include appropriate isotype control antibodies matched for species, class, and concentration to identify effects attributable to the Fc portion rather than IL-16 binding. Perform dose-response experiments—specific effects typically show a clear dose dependency while non-specific effects often appear at high concentrations without proportional increases at lower doses. Use multiple anti-IL-16 antibody clones targeting different epitopes (such as 14.1 and 17.1); effects observed with multiple antibodies are more likely to be specific. Complement antibody approaches with genetic manipulation strategies (siRNA, CRISPR) to knockdown IL-16 or its receptor—effects observed with both approaches strongly support specificity. Pre-absorb the antibody with recombinant IL-16 before adding to the experimental system; this should eliminate specific effects while non-specific effects would persist. For advanced validation, use F(ab')₂ or Fab fragments that lack the Fc region to eliminate Fc-mediated effects. Employ "rescue experiments" by adding excess recombinant IL-16 after antibody treatment—specific effects should be overcome with sufficient ligand concentration. Finally, conduct parallel experiments in systems known to lack IL-16 expression as biological negative controls to identify any antibody effects that occur independently of target binding .
The most appropriate statistical approaches for analyzing IL-16 antibody experimental data depend on the specific experimental design and data characteristics. For dose-response experiments testing antibody neutralization efficacy, nonlinear regression models should be used to determine EC50 values and confidence intervals, as demonstrated in the analysis of the 14.1 antibody binding to wild-type IL-16 versus the W600A variant. When comparing antibody effects across multiple experimental groups (e.g., different disease models or treatment conditions), analysis of variance (ANOVA) with appropriate post-hoc tests (Tukey's or Bonferroni) should be employed to account for multiple comparisons. For time-course experiments evaluating antibody effects, repeated measures ANOVA or mixed-effects models are more appropriate than multiple t-tests. When analyzing binding data from techniques like surface plasmon resonance or bio-layer interferometry, use association-dissociation kinetic models to determine kon, koff, and KD values. For ELISA data, four-parameter logistic regression typically provides better standard curve fitting than linear models. To assess agreement between different anti-IL-16 antibody clones or detection methods, use Bland-Altman analysis rather than simple correlation coefficients. When dealing with non-normally distributed data (common in biological systems), consider non-parametric alternatives such as Mann-Whitney or Kruskal-Wallis tests. For all analyses, report effect sizes and confidence intervals alongside p-values to better communicate biological significance beyond statistical significance .
The structural insights into IL-16-antibody interactions revealed by crystallographic and NMR studies open several promising avenues for next-generation therapeutics. The unexpected conformational change in the IL-16 PDZ domain induced by the 14.1 antibody binding—specifically the rotation of the αB-helix and movement of the Trp600 residue—provides a blueprint for designing small molecule inhibitors that could mimic this interaction. Rather than directly blocking the CD4 binding site, these molecules could allosterically modulate IL-16 function by stabilizing the conformationally altered state. The revelation of a cryptic peptide binding site that becomes accessible upon antibody binding suggests potential for developing peptide-based therapeutics that selectively target this site. Structure-based computational screening could identify compounds that bind specifically to the conformational epitope recognized by the 14.1 antibody. Additionally, the detailed characterization of the antibody-IL-16 interface, which buries 876 Ų of surface area with a mixture of polar and hydrophobic contacts, provides critical information for antibody engineering approaches such as affinity maturation or the development of bispecific antibodies that simultaneously target IL-16 and other inflammatory mediators. Understanding the key role of Trp600 in the binding interaction offers opportunities to design therapeutics that specifically exploit this structural feature for enhanced specificity and potency against IL-16-mediated inflammation .
Emerging technologies offer significant potential to enhance IL-16 targeting specificity and efficacy in both research and therapeutic contexts. Single-domain antibodies (nanobodies) derived from camelid antibodies could provide superior tissue penetration and access to conformational epitopes on IL-16 that are inaccessible to conventional antibodies. These smaller binding molecules might better access the critical conformational changes identified in the IL-16 PDZ domain. CRISPR-Cas9-based approaches for precise genome editing could enable the development of cell therapies with modified IL-16 signaling pathways, creating cells that are resistant to IL-16-mediated inflammation for adoptive transfer therapies. DNA/RNA aptamer technology presents opportunities to develop nucleic acid-based IL-16 binders with high specificity and potentially lower immunogenicity than protein therapeutics. Advanced antibody engineering techniques, including bispecific antibodies that simultaneously target IL-16 and CD4+ cells, could provide more targeted immunomodulation. Computational structural biology and artificial intelligence approaches will accelerate in silico screening of millions of compounds to identify those that specifically induce or stabilize the conformational changes observed with the 14.1 antibody binding. Antibody-drug conjugates could deliver anti-inflammatory compounds specifically to sites of IL-16 expression. Finally, improved delivery systems such as lipid nanoparticles could enhance the pharmacokinetics and tissue-specific targeting of anti-IL-16 therapeutics, particularly for conditions affecting tissues with specialized barriers like the central nervous system in multiple sclerosis .
Researchers can better integrate IL-16 pathway analysis into broader inflammatory disease models through several innovative approaches. Implement systems biology frameworks that simultaneously examine multiple inflammatory mediators alongside IL-16, using techniques like mass cytometry (CyTOF) or multi-parameter flow cytometry to correlate IL-16 signaling with the activation status of diverse immune cell populations. Develop transgenic reporter animal models expressing fluorescent proteins under IL-16 promoter control to visualize IL-16 expression patterns in real-time during disease progression. Combine single-cell RNA sequencing with spatial transcriptomics to map IL-16 expression and signaling at single-cell resolution within tissue microenvironments, revealing cell type-specific roles in disease pathology. Employ tissue-specific conditional knockout models for IL-16 or its receptor to dissect compartment-specific contributions to inflammatory conditions. Utilize multi-omics approaches integrating transcriptomics, proteomics, and metabolomics data to position IL-16 signaling within larger inflammatory networks, identifying key nodes and feedback mechanisms. Explore therapeutic combinations targeting IL-16 alongside other inflammatory pathways to address the complex redundancy in immune signaling. Develop organoid models incorporating IL-16-responsive immune cells to create more physiologically relevant disease models for drug screening. Finally, leverage artificial intelligence and machine learning approaches to analyze complex datasets and identify non-obvious relationships between IL-16 signaling and disease manifestations across different inflammatory conditions, potentially revealing new therapeutic opportunities .
IL-16 antibodies hold exceptional promise for precision medicine approaches to inflammatory diseases through several innovative applications. Patient stratification represents a primary opportunity—developing diagnostic assays measuring IL-16 levels or activity could identify patient subgroups with IL-16-driven pathology who would benefit most from targeted anti-IL-16 therapies. The cross-reactivity of antibodies like clone 14.1 with both human and mouse IL-16 facilitates translational research, allowing direct comparison between preclinical models and clinical applications. For conditions like rheumatoid arthritis, multiple sclerosis, and systemic lupus erythematosus where IL-16 levels are elevated, developing companion diagnostics that predict response to IL-16-targeting therapies could significantly improve treatment outcomes. The structure-based understanding of how the 14.1 antibody induces conformational changes in IL-16 opens possibilities for developing allosteric modulators rather than direct inhibitors, potentially offering more nuanced control of IL-16 activity. Tissue-specific delivery of anti-IL-16 antibodies using targeting moieties could enhance efficacy while reducing systemic effects. Combination therapies pairing IL-16 antibodies with existing biologics could address the complex, multi-factorial nature of inflammatory diseases. Sequential therapy approaches might use IL-16 antibodies during specific disease phases, such as acute flares, when IL-16 activity is highest. Finally, longitudinal monitoring of IL-16 levels during treatment could enable dynamic adjustment of therapeutic regimens, truly personalizing inflammatory disease management based on individual patient response patterns .
Interleukin-16 (IL-16), formerly known as lymphocyte chemoattractant factor (LCF), is a pleiotropic cytokine that plays multiple roles in the immune system. It is chemotactic for CD4+ T lymphocytes, monocytes, and eosinophils, meaning it can attract these cells to sites of inflammation . IL-16 can also upregulate the expression of IL-2 receptors and HLA-DR4, inhibit T cell receptor (TcR)/CD3-dependent activation, and repress HIV-1 transcription .
Structure and Processing: IL-16 is synthesized as a precursor molecule (pro-IL-16) of approximately 68 kDa and 631 amino acids (aa) lacking a signal peptide . The gene for IL-16 maps to chromosome 15 in humans and chromosome 7 in mice . The sequence and overall structure of IL-16 is conserved across species, with human and mouse IL-16 genes comprising seven exons and six introns . The human pro-IL-16 sequence displays over 90% similarity to various non-human primates .
In CD8+ T cells, active caspase-3 cleaves pro-IL-16, producing a biologically active, secreted form of IL-16, which represents 121 C-terminal aa residues of pro-IL-16 . The mechanism of release or secretion of IL-16 is currently unknown but does not appear to correlate with apoptosis .
Function: IL-16 functions as a chemoattractant, modulator of T cell activation, and inhibitor of HIV replication . It stimulates a migratory response in CD4+ lymphocytes, monocytes, and eosinophils, primes CD4+ T-cells for IL-2 and IL-15 responsiveness, and induces T-lymphocyte expression of the interleukin 2 receptor . IL-16 also serves as a ligand for CD4, initiating signaling pathways that increase intracytoplasmic calcium and inositol trisphosphate, activate p56lck, and translocate protein kinase C from the cytosol to the cell membrane .
Mouse Anti-Human IL-16 Antibody is a monoclonal antibody that specifically targets human IL-16. These antibodies are used in various research and clinical applications to study the role of IL-16 in immune responses and diseases.
Applications:
Mechanism of Action: The mouse anti-human IL-16 antibody binds to IL-16, preventing it from interacting with its receptor, CD4 . This inhibition blocks the downstream signaling pathways activated by IL-16, thereby reducing the recruitment and activation of immune cells .