Interleukin-16 (IL-16) is a pleiotropic cytokine with immunomodulatory roles in mice, primarily functioning as a chemoattractant for CD4+ T cells, monocytes, eosinophils, and dendritic cells . Produced as a precursor protein (pro-IL-16), it undergoes caspase-3-dependent processing to release its bioactive C-terminal peptide, which binds CD4 to mediate signaling . Mouse IL-16 shares 82.1% sequence similarity with human IL-16 in the C-terminal region, with conserved structural and functional properties .
Protein:
Property | Details | Source |
---|---|---|
Molecular Weight | 13.2 kDa (mature) | |
Purity (Recombinant) | ≥95% (SDS-PAGE) | |
Endotoxin Levels | ≤1 EU/μg | |
Reconstitution | Sterile water (0.1 mg/mL), 0.1% BSA |
Chemoattraction: Recruits CD4+ T cells, monocytes, and dendritic cells to inflammatory sites .
T Cell Activation: Upregulates CD25 (IL-2 receptor) and modulates Th1/Th2 balance .
Viral Suppression: Inhibits HIV and SIV replication in vitro .
Neutralizing IL-16 in NOD mice reduced insulitis and type 1 diabetes incidence by 50%, attributed to decreased CD4+ T-cell trafficking and CCL4-dependent mechanisms .
IL-16 mRNA peaks at 15 weeks in diabetic NOD mice, correlating with invasive insulitis .
Pro-Tumor Effects: IL-16 promotes lymphoma growth by increasing M2 macrophage infiltration, angiogenesis, and immunosuppressive cytokines (IL-6, IL-10) .
Anti-Tumor Effects: Exogenous IL-16 enhances CD8+ T-cell cytotoxicity and Th1 responses in breast cancer models, improving immunotherapy efficacy .
In contact hypersensitivity, IL-16 blockade reduced CD4+ T-cell infiltration but exacerbated CD8+ T-cell-mediated inflammation .
Anti-IL-16 antibody therapy in NOD mice:
IL-16 treatment in MMTV-PyMT breast cancer mice:
IL-16 is a chemoattractant cytokine that primarily recruits CD4-expressing cells in mouse models. Functionally, IL-16 plays a significant role in mediating T-cell responses in various inflammatory conditions, including contact hypersensitivity and viral infections. In mouse models of contact hypersensitivity, IL-16 is produced in both the epidermis and dermis during the elicitation phase of the immune response, with production beginning approximately 12 hours after challenge and increasing in a time-dependent manner for up to 72 hours . Interestingly, IL-16 demonstrates multifaceted roles in immune regulation, as it has been shown to have a down-regulating effect in contact hypersensitivity through CD4+ T cell recruitment, while enhancing susceptibility to influenza A virus infection through suppression of type I interferon responses . Unlike many other interleukins, IL-16 is produced by a variety of cells beyond traditional immune cells, making it a unique target for understanding immune cross-communication.
IL-16 can be measured in multiple biological samples from mice, allowing researchers to comprehensively assess its expression across different compartments. Serum represents one of the most accessible samples and can be obtained through standard blood collection methods and subsequent centrifugation to separate cellular components . Bronchoalveolar lavage fluid (BALF) offers insight into IL-16 levels specifically within the pulmonary microenvironment and can be acquired using a disposable syringe with a small-diameter needle inserted into the exposed trachea . Typically, sterile PBS is injected and aspirated back multiple times to maximize recovery of immune mediators. For tissue-specific analysis, organs such as lungs can be harvested, homogenized in appropriate media using an automatic tissue grinder, and clarified by centrifugation . Additionally, epidermal and dermal preparations from skin can be separately processed to determine compartment-specific expression in dermatological studies . Cell culture supernatants from ex vivo stimulated cells or tissues also serve as valuable samples for assessing IL-16 secretion in controlled experimental conditions .
The relationship between IL-16 and contact hypersensitivity (CHS) reveals a complex immunoregulatory role for this cytokine. Methodologically, researchers have established that examining both the sensitization and elicitation phases separately is critical for understanding IL-16's function in CHS. During the sensitization phase, a single application of haptens such as trinitrochlorobenzene (TNCB) and oxazolone induces IL-16 production, whereas primary irritants or vehicle controls do not trigger IL-16 expression . The kinetics of IL-16 production can be effectively studied through time-course experiments with tissue collection at intervals following hapten challenge, with ELISA analysis showing IL-16 becomes detectable at 12 hours post-challenge with levels progressively increasing up to 72 hours . Importantly, neutralizing antibody studies have provided critical insights into IL-16's regulatory function, as treatment of sensitized mice with anti-IL-16 neutralizing monoclonal antibodies enhances ear swelling responses while reducing the number of infiltrating CD4+ T cells . This seemingly paradoxical finding indicates that IL-16 recruits CD4+ T cells that subsequently exhibit down-regulating properties in the CHS response. To comprehensively study this relationship, researchers should employ a combination of approaches including immunohistochemistry to identify IL-16-producing cells, flow cytometry to characterize infiltrating immune cell populations, and functional blockade studies using neutralizing antibodies or genetic knockout models.
IL-16 enhances host susceptibility to influenza A virus (IAV) infection through multiple mechanisms, primarily involving the suppression of antiviral immune responses. Research methodologies have revealed that IL-16 does not affect viral binding or entry, as demonstrated by virus binding assays at 4°C and viral entry assays with time points at 15 minutes, 30 minutes, and 1 hour post-infection showing no significant differences between IL-16-expressing and control cells . Instead, IL-16 appears to function primarily by inhibiting type I interferon signaling. Mechanistic studies utilizing luciferase reporter assays have demonstrated that IL-16 expression markedly blocks IFN-β and ISRE (Interferon-Stimulated Response Element) activation . The suppression of this antiviral pathway is further evidenced by experiments showing that IL-16 deficiency dramatically elevates IFN-β and ISG15 (Interferon Stimulated Gene 15) expression after IAV infection in mouse embryonic fibroblasts . In vivo studies corroborate these findings, with significantly higher IFN-β mRNA and protein levels detected in lung homogenates and BALF samples from IL-16 knockout mice compared to wild-type mice at day 3 post-infection . These molecular interactions are best studied through a combination of approaches including gene expression analysis, protein interaction studies, signaling pathway activation assays, and targeted gene knockdown or knockout models to confirm the specific points at which IL-16 interferes with antiviral immunity.
IL-16 knockout (KO) mice display remarkable resistance to influenza A virus (IAV) infection compared to wild-type mice, providing crucial insights into IL-16's immunological functions. In experimental infection models using 8,000 PFU of PR8 influenza virus, wild-type mice exhibit rapid weight loss reaching 25-30% by days 8-10 post-infection, whereas IL-16 KO mice maintain normal body weight similar to mock-infected controls . Viral load assessment at day 7 post-infection reveals significantly reduced viral titers in the lungs of IL-16 KO mice compared to wild-type mice, demonstrating enhanced viral clearance in the absence of IL-16 . The mechanism underlying this protection appears multifaceted, involving both innate and adaptive immune components. At the cellular level, mouse embryonic fibroblasts (MEFs) derived from IL-16 KO mice show significantly reduced expression of viral proteins (NP, NS, and HA) and viral genes (NA, HA, and M) following IAV infection compared to wild-type MEFs . This resistance correlates with enhanced type I interferon responses, as IL-16 deficiency markedly increases IFN-β and ISG15 expression both before and after IAV infection . Beyond innate immunity, recent research indicates that IL-16 deficiency promotes Th1 and cytotoxic T lymphocyte (CTL) responses against IAV infection, suggesting that IL-16 normally restrains these protective adaptive immune mechanisms . These findings collectively indicate that IL-16 functions as a negative regulator of antiviral immunity at multiple levels, with implications for understanding immune evasion strategies employed by viruses and potential therapeutic approaches targeting IL-16.
Enzyme-linked immunosorbent assay (ELISA) represents the gold standard for quantifying IL-16 in mouse samples, offering sensitive and specific detection across multiple sample types. Commercial kits like the RayBio Mouse IL-16 ELISA Kit provide standardized protocols for measuring IL-16 in serum, plasma, and cell culture supernatants with high reproducibility . The ELISA methodology typically involves coating wells with capture antibody, incubating with samples and standards for 2 hours at room temperature, washing to remove unbound material, and then incubating with detection antibody for another 2 hours before adding substrate solution and measuring optical density at 450nm . For detecting IL-16 in tissue homogenates, supplementary sample preparation steps are crucial and typically involve homogenizing tissues in appropriate buffer using an automatic tissue grinder at 60Hz for 1 minute, followed by centrifugation at 8,000rpm for 10 minutes to obtain clarified supernatants . Immunoblot analysis provides an alternative approach for detecting IL-16 protein in mouse samples, particularly useful for confirming the presence of specific IL-16 isoforms or processing variants . For gene expression analysis, quantitative real-time PCR (qRT-PCR) with IL-16-specific primers allows researchers to measure IL-16 mRNA levels in cells and tissues, providing insight into transcriptional regulation under different experimental conditions. Flow cytometry can also be employed to detect intracellular IL-16 in specific cell populations, offering the advantage of simultaneous phenotypic characterization of IL-16-producing cells.
Generating and validating IL-16 knockout mouse models requires systematic approaches to ensure complete gene inactivation and exclude compensatory mechanisms. The CRISPR/Cas9 system has emerged as the predominant method for creating IL-16 knockout mice, offering precise genome editing capabilities. When designing guide RNAs, targeting early exons is recommended to ensure functional disruption of all protein isoforms and alternative splicing variants. Validation of gene deletion should employ multiple complementary techniques, beginning with genomic PCR to confirm the intended genetic alteration followed by reverse transcription PCR (RT-PCR) to verify absence of IL-16 mRNA expression in multiple tissues . Protein-level validation through immunoblot analysis represents a critical step, as demonstrated in studies where IL-16 knockout efficiency was confirmed by immunoblot analyses comparing wild-type and IL-16 KO mouse embryonic fibroblasts . Functional validation is equally important, assessing known IL-16-dependent processes such as CD4+ T cell recruitment in inflammatory models. Additionally, researchers should characterize baseline phenotypes of IL-16 knockout mice, paying particular attention to immune cell development and distribution, as IL-16 may have developmental roles beyond its immediate immunomodulatory functions. Potential compensatory mechanisms should be investigated by examining expression of functionally related cytokines and chemokines. Finally, confirmation of phenotypic rescue through reintroduction of IL-16 (via viral vectors or transgenic approaches) provides definitive evidence that observed phenotypes are specifically due to IL-16 deficiency rather than off-target effects.
Designing robust experiments to investigate IL-16's role in disease models requires careful consideration of temporal dynamics, tissue specificity, and mechanistic pathways. Initially, researchers should establish baseline IL-16 expression patterns in their disease model, as IL-16 is typically undetectable under steady-state conditions but increases following various immune challenges . Time-course studies are essential, as IL-16 production follows distinct kinetics depending on the model—appearing 12 hours after challenge in contact hypersensitivity and increasing for up to 72 hours . Multiple complementary approaches should be employed, including both gain-of-function (IL-16 overexpression) and loss-of-function (neutralizing antibodies, genetic knockout) strategies to comprehensively assess IL-16's contribution. For example, in influenza models, comparing IL-16 overexpression in A549 cells with IL-16 knockout in mouse embryonic fibroblasts provided consistent and complementary evidence for IL-16's role in enhancing viral infection . Cell-specific contributions should be delineated using techniques such as immunohistochemistry, which revealed that IL-16 is produced mainly by CD11c-negative cells in the epidermis during contact hypersensitivity . Researchers should consider both direct and indirect effects of IL-16, examining not only immediate cellular targets (CD4+ cells) but also downstream immunological consequences. For mechanistic insights, signaling pathway analysis is crucial—as demonstrated by studies showing IL-16's inhibitory effect on type I interferon responses during viral infection . Finally, translation between in vitro and in vivo findings must be verified, as cellular observations may not always predict whole-organism responses due to the complex interplay of multiple immune components.
Distinguishing between direct and indirect effects of IL-16 in inflammation models presents significant methodological challenges requiring sophisticated experimental designs. One fundamental challenge stems from IL-16's dual roles—it functions as both a chemoattractant for CD4+ cells and a modulator of cell signaling pathways. For example, in contact hypersensitivity, treatment with anti-IL-16 neutralizing antibodies paradoxically enhances ear swelling while reducing CD4+ T cell infiltration, suggesting that IL-16-recruited CD4+ T cells have downstream regulatory functions distinct from IL-16's direct chemoattractant effects . Temporal analysis becomes critical, as demonstrated in studies showing that initial IL-16 induction begins 12 hours after challenge, with effects manifesting over 72 hours, creating complex causality relationships that can be difficult to untangle . Cell-specific knockout or conditional expression systems offer valuable approaches to isolate IL-16's effects in particular cellular compartments, though interpreting results requires caution as deleting IL-16 from one cell type may alter the broader immune microenvironment. Adoptive transfer experiments combining cells from wild-type and IL-16 knockout mice can help delineate which aspects of inflammation depend on IL-16 production by specific cell populations versus IL-16 responsiveness in target cells. Ex vivo analysis of cells isolated from inflammatory sites allows assessment of direct IL-16 effects on cellular phenotypes, though these findings must be interpreted in the context of the complex in vivo environment. Finally, mechanistic studies examining signaling pathway activation, such as those demonstrating IL-16's inhibitory effect on type I interferon signaling during viral infection, provide molecular insights that help connect direct IL-16 interactions with downstream immune outcomes .
Effective measurement and interpretation of IL-16 production across tissues and cell types require integrating multiple complementary techniques with appropriate normalization strategies. Tissue-specific analysis starts with proper sample collection and processing—for bronchoalveolar lavage fluid, standardized protocols involving tracheal cannulation and repeated gentle lavage with fixed volumes of PBS ensure consistent recovery of airway cytokines . For solid tissues, homogenization procedures must be optimized for each tissue type, with lung tissues typically processed using automated tissue grinders at 60Hz for 1 minute, followed by clarification by centrifugation . When comparing IL-16 levels across tissues with different cellular compositions and densities, normalization to total protein content or tissue weight is essential. For cell-specific analysis, flow cytometry with intracellular cytokine staining allows identification of IL-16-producing cells within heterogeneous populations, though careful attention to stimulation conditions and transport inhibitors is required to capture accurate snapshots of IL-16 production. Single-cell RNA sequencing provides comprehensive insights into cell-specific IL-16 expression patterns and can reveal previously unrecognized producer populations. When interpreting results, researchers should consider that IL-16 exists as a precursor protein that requires processing to generate the active secreted form, necessitating analysis of both intracellular and secreted IL-16 to fully understand its regulation . Additionally, the detection threshold varies across techniques and tissues—for instance, baseline IL-16 is undetectable in normal skin but measurable in serum . Finally, researchers should remember that IL-16 production often exhibits strong temporal dynamics, as seen in contact hypersensitivity where levels gradually increase over 72 hours post-challenge, requiring thoughtful time-point selection to capture the full production profile .
IL-16 operates within a complex network of cytokines and immune mediators, with significant cross-talk that shapes inflammatory outcomes in mouse models. One critical interaction occurs between IL-16 and type I interferons, where IL-16 has been shown to markedly block IFN-β and ISRE activation, as demonstrated through luciferase reporter assays . This inhibitory relationship is bidirectional, as IL-16 deficiency dramatically elevates IFN-β and ISG15 expression following influenza A virus infection . Beyond interferons, IL-16 interacts with chemokines involved in T cell trafficking, potentially competing with or complementing their function depending on the inflammatory context. In contact hypersensitivity, IL-16 appears to work in concert with other chemoattractants to orchestrate the sequential recruitment of different immune cell populations, with IL-16 specifically targeting CD4+ T cells that subsequently exhibit regulatory properties . The temporal aspect of these interactions is crucial, as different cytokines dominate at distinct phases of the inflammatory response—IL-16 production begins 12 hours after challenge in contact hypersensitivity and increases over 72 hours, likely intersecting with changing cytokine milieus throughout this period . Additionally, IL-16 may influence cytokine production by target cells, as suggested by its effect on T helper cell polarization, where IL-16 deficiency promotes Th1 responses during viral infection . These complex interactions can be studied through multiplex cytokine analysis of inflammatory sites, correlation analysis between IL-16 and other mediators across disease time courses, and ex vivo stimulation of cells with IL-16 to assess changes in their cytokine production profiles.
IL-16 exhibits complex and context-dependent relationships with CD4+ T cell subsets across different mouse disease models, influencing both cell recruitment and functional polarization. In contact hypersensitivity, IL-16 selectively recruits CD4+ T cells that subsequently demonstrate immunoregulatory properties, as evidenced by experiments where anti-IL-16 neutralizing antibody treatment reduced CD4+ T cell infiltration while paradoxically enhancing ear swelling responses . This suggests IL-16 specifically attracts regulatory T cell subsets or induces regulatory functions in recruited cells. Conversely, in influenza A virus infection models, IL-16 deficiency promotes Th1 responses, indicating that IL-16 normally constrains this CD4+ T cell subset . This dual nature of IL-16—recruiting CD4+ T cells while potentially modulating their functional differentiation—creates complex outcomes in different disease contexts. Methodologically, characterizing this relationship requires multiparameter flow cytometry to simultaneously assess T cell subset markers (Th1, Th2, Th17, Treg) along with activation status and cytokine production potential. Adoptive transfer experiments using labeled CD4+ T cell subsets can track their differential recruitment in response to IL-16, while conditional knockout approaches can evaluate how IL-16 deficiency in specific cellular compartments affects T helper cell polarization. Additionally, ex vivo stimulation of isolated CD4+ T cell subsets with recombinant IL-16 can reveal direct effects on their activation, proliferation, and cytokine production profiles. Finally, temporal analysis is crucial, as IL-16's effects on T cell recruitment and differentiation may evolve throughout disease progression, requiring assessment at multiple time points to capture the dynamic nature of this relationship.
The absence of IL-16, as studied in knockout mouse models, profoundly affects both innate and adaptive immune responses, often resulting in enhanced protection against certain pathogens. In the context of innate immunity, IL-16 deficiency significantly amplifies type I interferon responses, with IL-16 knockout mouse embryonic fibroblasts showing elevated baseline levels of IFN-β and ISG15, which become dramatically increased following influenza A virus infection . This enhanced antiviral state translates to the whole organism level, where IL-16 knockout mice display significantly higher IFN-β mRNA and protein levels in lung homogenates and bronchoalveolar lavage fluid on day 3 post-influenza infection compared to wild-type mice . The mechanism appears to involve direct inhibition of interferon signaling by IL-16, as demonstrated by experiments showing IL-16 expression markedly blocks IFN-β and ISRE activation in reporter assays . On the adaptive immunity front, IL-16 deficiency promotes Th1 and cytotoxic T lymphocyte (CTL) responses against influenza A virus infection, suggesting IL-16 normally restrains these protective adaptive immune mechanisms . This enhanced cell-mediated immunity likely works in concert with elevated type I interferon responses, as IFN-α/β signaling is known to promote Th1 responses and provide the "third signal" needed by naive CD8+ T cells responding to pathogens . The functional consequence of these immunological changes is dramatically improved resistance to influenza A virus challenge, with IL-16 knockout mice maintaining normal body weight following infection with 8,000 PFU of PR8 virus, while wild-type mice lose 25-30% of their body weight . These findings collectively indicate that IL-16 serves as a negative regulator of both innate and adaptive antiviral immunity, with its absence unleashing more effective immune protection.
While IL-16 has been extensively studied in peripheral inflammatory conditions, its roles in neuroinflammatory and neurodegenerative mouse models represent an emerging frontier with significant therapeutic implications. Beyond its established function as a chemoattractant cytokine in peripheral tissues, IL-16 has unique properties in the central nervous system (CNS) where it can be produced by both resident glial cells and infiltrating immune cells during inflammatory conditions. The methodological approaches for studying IL-16 in the CNS require specialized techniques, including cerebrospinal fluid sampling, brain region-specific tissue processing, and primary glial cell cultures from mouse brains. Researchers investigating IL-16 in neuroinflammation should employ immunohistochemistry with cell-type specific markers to identify the cellular sources of IL-16 within the CNS, particularly during pathological states. Functional studies using IL-16 knockout mice in models of multiple sclerosis (experimental autoimmune encephalomyelitis), Alzheimer's disease, Parkinson's disease, and traumatic brain injury can reveal condition-specific roles for this cytokine. The blood-brain barrier presents a unique consideration in neuroinflammatory studies, requiring researchers to distinguish between infiltrating peripheral immune cells responding to IL-16 gradients versus local IL-16 production and signaling within the CNS parenchyma. Given IL-16's ability to inhibit type I interferon responses in peripheral tissues as observed in influenza infection models, similar mechanisms may operate in viral encephalitis, where type I interferons play crucial protective roles . Additionally, IL-16's capacity to modulate CD4+ T cell responses could significantly impact the pathogenesis of T cell-mediated neuroinflammatory conditions, potentially offering new therapeutic targets for these often-intractable diseases.
The contribution of IL-16 to immunosenescence represents an important but understudied area that may provide insights into age-related disease susceptibility. Methodologically, investigating age-related changes in IL-16 requires carefully designed longitudinal studies comparing young (8-12 weeks), middle-aged (12-15 months), and aged (18-24 months) mice, with age-matched IL-16 knockout controls to determine causality versus correlation. Researchers should examine both basal IL-16 expression across tissues and its inducibility following immune challenges, as aging often affects not just baseline cytokine levels but also their responsiveness to stimulation. Flow cytometric analysis of IL-16 receptor (CD4) expression on various immune cell populations throughout the lifespan can reveal potential changes in cellular responsiveness to IL-16 signaling. Given IL-16's role in modulating CD4+ T cell responses and its inhibitory effect on type I interferon signaling, age-related alterations in these functions may contribute to the well-documented decline in T cell responses and antiviral immunity in elderly populations . The enhanced antiviral resistance observed in IL-16 knockout mice during influenza infection raises the intriguing possibility that IL-16 blockade might partially restore impaired antiviral immunity in aged mice . Additionally, since IL-16 deficiency promotes Th1 and cytotoxic T lymphocyte responses, therapeutic targeting of IL-16 might help counteract the age-related shift toward Th2 responses that contributes to declining cell-mediated immunity . Beyond infectious challenges, researchers should investigate IL-16's role in inflammaging—the chronic, low-grade inflammation characteristic of advanced age—by measuring IL-16 levels in various tissues throughout the lifespan and correlating these with markers of cellular senescence and inflammatory mediators.
Reconciling contradictory findings about IL-16 across disease models requires methodological rigor and contextual interpretation that accounts for multiple variables. The most striking contradiction appears between contact hypersensitivity models, where IL-16 exhibits immunoregulatory functions through recruiting CD4+ T cells with down-regulatory properties, and influenza infection models, where IL-16 enhances susceptibility by suppressing antiviral immunity . These apparent contradictions can be systematically addressed through several approaches. First, careful examination of the disease-specific immune microenvironment is essential, as IL-16 may interact with different cytokine networks in each condition, producing context-dependent outcomes. Second, the kinetics of IL-16 production and action differ substantially between acute viral infections and delayed-type hypersensitivity reactions, potentially explaining divergent functions at different stages of immune responses. Third, cell-specific effects must be considered—IL-16 may recruit regulatory CD4+ T cells in skin inflammation while primarily affecting type I interferon-producing cells in viral infections . Researchers should employ identical reagents and protocols when directly comparing IL-16 functions across models to minimize technical variables. Additionally, simultaneous examination of multiple readouts (cellular infiltration, molecular signaling, functional outcomes) provides more comprehensive understanding than single-parameter analyses. Genetic background differences between studies can significantly impact cytokine functions, necessitating standardization or explicit acknowledgment of strain differences. Finally, researchers should consider that observed contradictions may reflect genuine biological complexity rather than experimental error, as cytokines frequently demonstrate pleiotropic and context-dependent functions in the immune system.
Current mouse models for studying IL-16 present several important limitations that researchers must consider when extrapolating findings to human diseases. A fundamental limitation involves species-specific differences in IL-16 biology—while human and mouse IL-16 share considerable sequence homology, they exhibit distinct expression patterns, processing requirements, and receptor interactions. The conventional IL-16 knockout mouse models typically eliminate all IL-16 isoforms throughout development, potentially triggering compensatory mechanisms that mask physiological functions of IL-16 in adult animals . Conditional and inducible knockout systems represent improvements but are still imperfect approximations of therapeutic IL-16 inhibition. Additionally, laboratory mice maintained in specific-pathogen-free facilities have immature immune systems compared to their wild counterparts or humans, potentially altering IL-16's immunoregulatory functions. The genetic homogeneity of inbred mouse strains contrasts sharply with human genetic diversity, limiting the generalizability of findings—for example, C57BL/6 mice used in many IL-16 studies have characteristic Th1-biased immune responses that may uniquely interact with IL-16 functions . Disease models often represent accelerated and simplified versions of complex human conditions, failing to capture chronic inflammatory processes where IL-16 may play distinct roles. Methodologically, detection assays for mouse IL-16 may have different sensitivities than those for human IL-16, complicating direct comparisons of expression levels . Finally, translational challenges arise from differences in drug pharmacokinetics and target tissue accessibility between mice and humans, affecting the performance of IL-16-targeting therapeutics. Despite these limitations, mouse models remain valuable when interpreted cautiously and complemented with human translational studies.
IL-16 is synthesized as a large, inactive precursor protein known as pro-IL-16. This precursor protein is approximately 80 kDa in size and is stored inside the cell until it is activated . The activation of pro-IL-16 involves proteolytic cleavage mediated by caspase-3, which results in the release of two functional proteins . The cytokine function of IL-16 is attributed to the secreted C-terminal region, which is about 14 kDa in size . The N-terminal product may play a role in cell cycle control .
IL-16 functions as a chemoattractant, attracting immune cells to sites of inflammation . It also modulates T cell activation and acts as an inhibitor of HIV replication . The signaling process of IL-16 is mediated by the CD4 receptor . This cytokine is involved in various immune responses and has been associated with the development of multiple sclerosis, an inflammatory demyelinating disease of the central nervous system .
Recombinant IL-16 is produced using various expression systems, including HEK293 cells and E. coli . The recombinant protein is purified to ensure high purity and low endotoxin levels, making it suitable for research applications . Recombinant mouse IL-16 is a non-glycosylated protein containing 127 amino acids and has a molecular weight of approximately 13.2 kDa .