CXCL16 belongs to the CXC chemokine family and is distinguished by its transmembrane domain, enabling dual expression as a cell-surface molecule and soluble chemokine. The murine CXCL16 precursor (246 amino acids) includes:
Chemokine domain (88 aa): Mediates receptor binding (CXCR6/Bonzo) .
Transmembrane domain (22 aa): Anchors the molecule to cell membranes .
Cytoplasmic tail (23 aa): Contains phosphorylation sites for intracellular signaling .
CXCL16 is also expressed in non-lymphoid tissues (e.g., lung, kidney) and induced by inflammatory cytokines like TNF-α and IFN-γ .
CXCL16 modulates immune cell recruitment and polarization:
CXCR6+ T cells/NKT cells: Directed to lymphoid organs and inflammatory sites .
Macrophage phenotype: Promotes anti-inflammatory polarization (ARG1↑, CD163↑) while suppressing pro-inflammatory genes (IL-1β↓, TNF-α↓) .
Apoptotic cell clearance: Binds phosphatidylserine but does not enhance phagocytosis .
CXCL16 critically regulates regenerative processes:
Muscle Regeneration: Deficiency in CXCL16−/− mice delays satellite cell differentiation (MyoD↓, myogenin↓) and increases fibrosis (TGF-β1↑) .
Cardiac Injury: CXCL16 deficiency improves survival post-myocardial infarction (MI) by reducing Ly6C<sup>high</sup> monocyte infiltration .
CXCL16 modulates synaptic transmission in the hippocampus:
Inhibitory Transmission: ↑ mIPSC frequency via GABA release .
Excitatory Transmission: ↑ mEPSC frequency and ↓ PPR, enhancing glutamate release .
Mechanism: Requires A3 adenosine receptors and microglial activity .
CXCL16 undergoes alternative splicing to generate isoforms with distinct functions:
CXCL16 is commercially available for research:
Product | Source/Tag | Purity | Applications |
---|---|---|---|
503-CX | E. coli (Asn27-Pro114) | >95% | Angiogenesis assays, receptor binding studies |
CXC-MM216 | HEK293 (C-hFc tag) | >95% | In vivo tumor models, fibrosis studies |
CXCL16 targeting has shown promise in preclinical models:
Mouse CXCL16 exhibits a specific expression pattern across multiple tissue types. It is prominently produced by dendritic cells in lymphoid organ T cell zones and by cells in the splenic red pulp in both membrane-bound and soluble forms. Northern blot analysis has identified significant expression in several non-lymphoid tissues including lung, small intestine, and kidney . In pathological conditions, expression increases in experimental and human nephropathies, with localization in parenchymal renal cells, vascular wall cells, leukocytes, and platelets . Within the central nervous system, CXCL16 is normally expressed and plays roles in neuroprotection against glutamate-induced damage through interactions with astrocytes, involving adenosine receptor type 3 (A3R) and the chemokine CCL2 . This diverse expression pattern highlights CXCL16's multifunctional nature in different physiological and pathological contexts.
The primary receptor for CXCL16 is CXCR6/Bonzo (also known as STRL33 and TYMSTR). This receptor demonstrates a selective expression pattern predominantly on naive CD8 cells, natural-killer T cells, and activated CD8 and CD4 T cells . Beyond immune cells, CXCR6 is prominently expressed on human endothelial progenitor cells (EPCs) and human microvascular endothelial cells (HMVECs), where its expression can be upregulated by pro-inflammatory cytokines such as interleukin-1β (IL-1β) . Interestingly, although HMVECs and EPCs both express CXCR6 and respond to CXCL16 stimulation, research has identified that they utilize distinct signal transduction pathways, suggesting cell type-specific mechanisms downstream of this receptor . The recognition of CXCR6's expression on endothelial cells has expanded our understanding of CXCL16's roles beyond immune cell trafficking to include vascular functions.
CXCL16 exerts multifaceted effects on neurotransmission in the mouse hippocampus through both GABAergic and glutamatergic pathways. Electrophysiological studies in acute hippocampal slices demonstrate that CXCL16 increases the frequency of miniature inhibitory postsynaptic currents (mIPSCs) without affecting their amplitude (CTRL: 2.71 ± 0.17 Hz, CXCL16: 3.52 ± 0.36 Hz, p = 0.008). This is evidenced by a leftward shift in the cumulative probability plot for inter-event intervals (IEI), suggesting presynaptic enhancement of spontaneous GABA release .
Paradoxically, CXCL16 reduces the amplitude of evoked IPSCs (eIPSCs) to approximately 72.7 ± 8% of control while increasing the paired-pulse ratio (PPR) from 1.67 ± 0.30 to 2.43 ± 0.48 (p = 0.03), indicating that it simultaneously reduces action potential-dependent GABA release. For glutamatergic transmission, CXCL16 increases miniature excitatory postsynaptic current (mEPSC) frequency (CTRL: 0.62 ± 0.13 Hz, CXCL16: 0.85 ± 0.13 Hz, p = 0.05) with no effect on amplitude, while enhancing evoked EPSC amplitude (CTRL: 224.69 ± 48.45 pA, CXCL16: 258.24 ± 47.88 pA, p = 0.025) and reducing PPR (CTRL: 1.41 ± 0.11, CXCL16: 1.26 ± 0.08, p = 0.025), suggesting increased glutamate release probability .
The modulatory effects of CXCL16 on synaptic transmission involve a complex interplay of cellular and molecular components. Research has established that CXCL16's actions require functional adenosine receptor type 3 (A3R), as these effects are abolished in A3R knockout mice. Microglia also serve as key mediators in this pathway, demonstrated by the loss of CXCL16's effects in wild-type slices treated with minocycline, a microglial inhibitor .
The differential effects on evoked versus spontaneous GABAergic transmission likely involve metabotropic GABA-B receptors, which specifically reduce evoked GABA release onto CA1 pyramidal neurons. Mechanistically, CXCL16 appears to function through cross-communication with astrocytes, involving A3R and possibly the chemokine CCL2, establishing a neuro-glial-immune signaling network that modulates neurotransmitter release at the synaptic level . These findings demonstrate that CXCL16's neuronal effects extend beyond direct receptor-mediated signaling to include complex multicellular interactions that ultimately alter synaptic function through both pre- and post-synaptic mechanisms.
Multiple lines of evidence establish CXCL16 as a potent angiogenic mediator in mouse models. In vitro studies demonstrate that CXCL16 induces capillary tube formation in endothelial cells, with quantitative assessment showing significant increases in nodular contacts between at least three endothelial cell tubes and circular tube network formations . CXCL16 also promotes dose-dependent migration of human microvascular endothelial cells (HMVECs) in modified Boyden chemotaxis systems, providing a mechanistic basis for its angiogenic effects .
In vivo evidence comes from the SCID mouse chimera system, where CXCL16 intragraft injection into normal synovial tissue resulted in HMVEC integration into engrafted synovium, not as scattered cells but as organized clumps forming vessels and chimeric nodes with surrounding murine vasculature within 48 hours. Furthermore, depletion of CXCL16 from rheumatoid arthritis synovial fluid reduced endothelial progenitor cell (EPC) recruitment by approximately two-thirds, confirming CXCL16's direct role in recruiting angiogenic precursors . These findings collectively establish CXCL16 as a central mediator of pathological angiogenesis, particularly in inflammatory disease contexts.
CXCL16-CXCR6 signaling plays a critical role in inflammatory arthritis development through multiple mechanisms. In the K/BxN serum-induced inflammatory arthritis model, CXCR6-deficient mice (CXCR6-/-) demonstrate profound reductions in hemoglobin levels that correlate with decreased monocyte and T-cell recruitment to arthritic joint tissues compared to wild-type mice . This suggests that CXCL16-CXCR6 interactions are essential for immune cell trafficking into inflamed joints.
Additionally, previous studies have shown that mice depleted of CXCL16 exhibit reduced arthritis severity, further supporting this chemokine's pathogenic role . Mechanistically, CXCL16 and CXCR6 appear central to endothelial progenitor cell recruitment and blood vessel formation in rheumatoid arthritis joints. Both TNF-α and CXCL16 can upregulate CXCL16 expression in synovial tissue, creating a positive feedback loop that amplifies inflammatory responses . The correlation between circulating EPC numbers and Disease Activity Scores in rheumatoid arthritis patients further suggests that CXCL16-mediated EPC recruitment contributes significantly to synovial vasculogenesis and disease progression, making this pathway a potential therapeutic target.
Several mouse models have proven valuable for investigating CXCL16 functions in vivo, each offering distinct advantages depending on the research question. The severe combined immunodeficient (SCID) mouse chimera system, where human synovial tissue is engrafted into SCID mice, provides an excellent platform for examining human endothelial cell and endothelial progenitor cell recruitment in response to CXCL16. This model allows for direct intragraft injection of CXCL16 or selective immunodepletion of CXCL16 from biological fluids, enabling precise manipulation of this signaling pathway in a humanized context .
For inflammatory arthritis studies, the K/BxN serum-induced arthritis model in CXCR6-deficient (CXCR6-/-) mice compared to wild-type C57BL/6 mice offers insights into the receptor-mediated effects of CXCL16. This model provides quantifiable endpoints including hemoglobin levels and immune cell recruitment that correlate with disease severity . For investigating neuronal functions, acute hippocampal slice preparations from both wild-type and A3R knockout mice treated with CXCL16, combined with electrophysiological recording techniques, have successfully delineated CXCL16's effects on neurotransmission . The availability of both ligand-depleted and receptor-deficient models provides complementary approaches to understand CXCL16 biology across multiple physiological systems.
Multiple complementary techniques have proven effective for assessing CXCL16-induced angiogenesis across different experimental contexts. In vitro, the capillary morphogenesis assay provides quantitative assessment of tube formation, measured by counting nodular contacts between at least three endothelial cell tubes and circular tube network formations in response to CXCL16 treatment . The modified Boyden chemotaxis system offers quantitative measurement of endothelial cell migration toward CXCL16 concentration gradients, providing insight into the directional migration component of angiogenesis .
For in vivo assessment, the SCID mouse chimera system combined with fluorescently labeled human microvascular endothelial cells (HMVECs) or endothelial progenitor cells (EPCs) allows visualization and quantification of cell recruitment and integration into developing vasculature. Immunofluorescence staining with anti-mouse/human von Willebrand factor (vWF) enables identification of chimeric vessels containing both mouse and human endothelial cells . In inflammatory arthritis models, measuring hemoglobin levels in joint tissue provides an indirect but quantifiable measure of vascular density that correlates with angiogenic activity . Collectively, these methods provide a comprehensive toolkit for examining CXCL16's angiogenic effects from molecular and cellular levels to intact tissue systems.
Several apparent contradictions emerge when examining CXCL16 functions across different experimental systems. One notable contradiction involves CXCL16's effects on GABAergic neurotransmission, where it simultaneously increases spontaneous GABA release (increased mIPSC frequency) but decreases evoked GABA release (reduced eIPSC amplitude) . This apparent paradox likely reflects distinct presynaptic mechanisms regulating action potential-dependent versus independent neurotransmitter release, possibly involving metabotropic GABA-B receptor activation that specifically suppresses evoked GABA release.
Another contradiction emerges when comparing CXCL16's protective roles in some contexts against its pathogenic functions in others. CXCL16 exerts neuroprotective effects against glutamate-induced damage in the CNS , while promoting inflammatory pathology in rheumatoid arthritis . Similarly, targeting CXCL16 protects against experimental glomerular injury or interstitial fibrosis , yet CXCL16 is required for proper immune function. These seemingly contradictory findings likely reflect tissue-specific and context-dependent roles of CXCL16-CXCR6 signaling. Resolution of these contradictions requires consideration of the local cellular microenvironment, temporal dynamics of CXCL16 signaling, and integration with other inflammatory and homeostatic pathways that may shift CXCL16 from protective to pathogenic functions depending on the biological context.
Studying the distinct biological activities of membrane-bound versus soluble CXCL16 presents several methodological challenges. Mouse CXCL16 exists in both forms, with the membrane-bound version functioning primarily as an adhesion molecule and scavenger receptor, while the soluble form acts as a chemokine following proteolytic cleavage . Distinguishing between these forms requires specialized techniques.
For membrane-bound CXCL16, surface biotinylation assays, flow cytometry with non-permeabilized cells, and immunofluorescence microscopy on non-permeabilized cells can specifically detect cell surface expression. To study cleavage mechanisms, inhibitors of metalloproteinases (particularly ADAM10 and ADAM17) can block the conversion to soluble form. For soluble CXCL16, ELISA assays with antibodies specific to regions absent in the membrane-bound form provide quantitative measurement in biological fluids.
Functionally separating the activities of these forms requires creative experimental approaches. Recombinant proteins lacking the transmembrane domain mimic soluble CXCL16, while mutants resistant to proteolytic cleavage can isolate membrane-bound functions. Cell-specific conditional knockout models that maintain expression in some tissues while deleting in others help delineate tissue-specific contributions. Understanding which form predominates in different pathological contexts remains challenging but essential for developing targeted therapeutic approaches that selectively modulate specific CXCL16 functions.
Targeting the CXCL16-CXCR6 axis therapeutically requires careful consideration of its diverse physiological roles. In inflammatory arthritis models, evidence suggests that inhibiting this pathway could provide therapeutic benefit, as CXCR6-deficient mice show reduced arthritis severity with decreased angiogenesis and immune cell recruitment . Similarly, targeting CXCL16 protected against experimental glomerular injury and interstitial fibrosis in kidney disease models , suggesting potential applications in nephropathies.
A nuanced therapeutic approach might involve tissue-specific delivery of inhibitors, temporal restriction of treatment to acute disease phases, or selective targeting of pathological functions while preserving homeostatic roles. Developing agents that specifically block the angiogenic or inflammatory functions of CXCL16 without affecting its scavenger receptor or neuroprotective activities represents an ideal but challenging approach requiring deeper understanding of structure-function relationships in this multifaceted molecule.
Several cutting-edge experimental approaches hold promise for addressing key knowledge gaps in CXCL16 biology. Single-cell RNA sequencing of tissues from normal and disease states could reveal cell-specific expression patterns and help identify novel cellular sources and targets of CXCL16. This approach would be particularly valuable for understanding the cellular networks involved in CXCL16 production and response during inflammation.
Structure-function studies using domain-specific mutations or chimeric proteins could delineate which regions of CXCL16 mediate its diverse functions, potentially enabling development of function-selective modulators. Advanced imaging techniques such as intravital microscopy in reporter mice expressing fluorescently tagged CXCL16 or CXCR6 would allow real-time visualization of ligand-receptor dynamics in living tissues during disease progression.
Conditional and inducible knockout models offer advantages over conventional knockouts by allowing temporal and spatial control of CXCL16 or CXCR6 deletion, helping distinguish developmental versus acute roles and tissue-specific functions. CRISPR-Cas9 genome editing could generate knockin mice expressing tagged versions of endogenous CXCL16 to track its production, processing, and distribution without overexpression artifacts. Combined with phosphoproteomics and interactome analysis, these approaches would provide comprehensive insights into CXCL16 signaling networks across different physiological contexts, potentially revealing new therapeutic targets within this pathway.
Designing rigorous experiments to study CXCL16 function requires multiple levels of controls. For specificity controls, comparing wild-type mice with CXCR6-knockout mice is essential to confirm receptor dependency of observed effects . Similarly, immunodepletion of CXCL16 from biological samples with validation by ELISA, compared to sham-depleted samples, confirms ligand-specific effects. Structure-function controls using heat-inactivated CXCL16 or irrelevant chemokines of similar size help establish specificity of biological responses.
Pathway validation controls should include inhibitors of known downstream signaling components (e.g., A3R antagonists when studying neurotransmission effects ) and cell-specific inhibitors (e.g., minocycline for microglial contribution). For studies involving recombinant CXCL16, expression system controls comparing protein from different sources (e.g., HEK293 versus E. coli) help exclude effects of post-translational modifications or contaminants.
Temporal controls establishing baseline measurements before intervention and appropriate time-course analyses are crucial given CXCL16's dynamic regulation. When studying disease models, both prophylactic (before disease onset) and therapeutic (after disease establishment) intervention timings should be tested. Finally, sex-balanced experimental groups are essential as sex differences in chemokine biology are increasingly recognized, potentially affecting the magnitude or even direction of CXCL16-mediated effects in different disease contexts.
When confronted with contradictory data on CXCL16 function, researchers should systematically evaluate several factors that might explain discrepancies. Context dependency is paramount—CXCL16 functions differently in distinct tissue microenvironments due to tissue-specific co-receptors, proteases, and downstream signaling machinery. The developmental or disease stage must also be considered, as CXCL16's role may shift from beneficial to detrimental as inflammation progresses from acute to chronic phases.
Technical variables can drive apparent contradictions, including differences in CXCL16 concentration between studies (physiological nanomolar ranges versus supraphysiological doses), soluble versus membrane-bound forms being studied, or variations in experimental readouts measuring different biological processes. Species differences (mouse versus human CXCL16) may also explain conflicting results, particularly in translational studies.
CXCL16 expression is regulated by multiple factors across different mouse tissues. Inflammatory cytokines are primary regulators, with TNF-α demonstrated to upregulate CXCL16 expression in synovial tissue in the SCID mouse chimera system . CXCL16 can also upregulate its own expression, suggesting positive feedback regulation that may amplify initial inflammatory signals . In vascular and renal cells, expression increases in response to tissue injury and during nephropathies , indicating stress-responsive regulation.
These regulatory pathways can be experimentally manipulated through several approaches. Cytokine stimulation using recombinant TNF-α, IFN-γ, or IL-1β in both in vitro and in vivo systems can induce CXCL16 expression. Conversely, cytokine neutralizing antibodies or receptor antagonists can block endogenous upregulation. Genetic approaches using promoter-reporter constructs can identify transcription factor binding sites regulating CXCL16 expression, while chromatin immunoprecipitation (ChIP) assays can confirm direct transcriptional regulation.
For tissue-specific manipulation, Cre-lox technology enabling conditional deletion or overexpression of CXCL16 in specific cell types provides powerful tools for dissecting tissue-specific functions. Viral vector-mediated local delivery of CXCL16 expression constructs or shRNA for knockdown offers an alternative approach for spatial control. Finally, environmental manipulations such as hypoxia, oxidative stress, or lipopolysaccharide challenge can be used to study physiological triggers of CXCL16 upregulation in different disease contexts.
Multiple complementary methods provide sensitive and specific detection of mouse CXCL16 across experimental samples. For protein detection in biological fluids, enzyme-linked immunosorbent assay (ELISA) offers quantitative measurement with high sensitivity, ideally using antibody pairs recognizing different epitopes to enhance specificity. Commercial mouse CXCL16 ELISA kits typically have detection limits in the low picogram/mL range, suitable for most physiological and pathological contexts.
In tissue samples, immunohistochemistry or immunofluorescence with validated antibodies provides spatial information about CXCL16 distribution and cellular sources. Double staining with cell type-specific markers can identify producing cells, while differential staining under permeabilized versus non-permeabilized conditions can distinguish membrane-bound from intracellular CXCL16. Flow cytometry offers an alternative for quantifying cell surface and intracellular CXCL16 in dissociated tissues or cultured cells.
For mRNA detection, quantitative RT-PCR provides sensitive measurement of CXCL16 transcript levels, while in situ hybridization localizes expression in intact tissues. RNA-sequencing approaches offer comprehensive analysis of CXCL16 expression alongside the entire transcriptome, revealing co-regulated genes. For functional assessment, bioassays measuring CXCR6-expressing reporter cell migration or calcium flux in response to samples provide a functional readout that confirms biological activity of the detected CXCL16, complementing pure quantitation methods.
Researchers investigating CXCL16-CXCR6 interactions have access to several specialized resources. Genetically modified mouse strains include CXCR6-deficient (CXCR6-/-) mice on C57BL/6 background for studying receptor-dependent functions and CXCR6-GFP reporter mice where GFP replaces one CXCR6 allele, allowing visualization of receptor-expressing cells. While complete CXCL16 knockout mice have limited availability, conditional floxed alleles are being developed for tissue-specific deletion studies.
Recombinant proteins include purified mouse CXCL16 expressed in various systems (HEK293, E. coli) with different tags (hFc, His, GST) for functional studies . Function-blocking antibodies against mouse CXCL16 or CXCR6 enable acute inhibition experiments without genetic compensation effects of knockouts. For detection, validated antibody pairs for ELISA, flow cytometry, and immunohistochemistry are commercially available with confirmed specificity.
Cell lines engineered to express mouse CXCR6 with various reporters (GFP, luciferase) facilitate high-throughput screening of CXCL16 function or inhibitor identification. Disease models with established CXCL16 involvement include the K/BxN serum-induced arthritis model , nephropathy models , and the SCID mouse chimera system for studying human cell interactions within mouse tissues . These resources collectively provide a robust toolkit for investigating CXCL16-CXCR6 biology across multiple experimental contexts.
Several electrophysiological techniques have proven effective for investigating CXCL16's modulatory effects on neurotransmission. Whole-cell patch-clamp recording in acute hippocampal slices provides direct measurement of synaptic currents, allowing detection of both miniature postsynaptic currents (mIPSCs, mEPSCs) that reflect spontaneous neurotransmitter release and evoked postsynaptic currents (eIPSCs, eEPSCs) measuring action potential-dependent release . This technique has successfully documented CXCL16's differential effects on GABAergic and glutamatergic transmission.
Paired-pulse stimulation protocols, where two stimuli are delivered at short intervals (e.g., 50ms), calculate the paired-pulse ratio (PPR) as a measure of presynaptic release probability. CXCL16 increases PPR for inhibitory transmission (from 1.67 ± 0.30 to 2.43 ± 0.48) while decreasing it for excitatory transmission (from 1.41 ± 0.11 to 1.26 ± 0.08), revealing pathway-specific presynaptic modulation .
Analysis of miniature postsynaptic current frequency and amplitude provides insight into pre- versus postsynaptic mechanisms, with frequency changes (as observed with CXCL16) typically indicating presynaptic modulation. Cumulative probability plots of inter-event intervals offer statistical power for detecting subtle changes in release probability. For mechanistic dissection, pharmacological tools including receptor antagonists (A3R antagonists), signaling pathway inhibitors, and glial modulators (minocycline) can be combined with electrophysiology to identify cellular and molecular components of CXCL16's neuromodulatory actions . These approaches collectively provide a comprehensive toolkit for characterizing CXCL16's effects on synaptic function.
Single-cell technologies offer transformative potential for understanding CXCL16-CXCR6 biology across multiple dimensions. Single-cell RNA sequencing (scRNA-seq) can identify the complete repertoire of cell types expressing CXCL16 and CXCR6 across tissues, potentially revealing previously unrecognized cellular sources and targets. This approach is particularly valuable for heterogeneous tissues like brain, spleen, and inflamed synovium where bulk analysis masks cell type-specific expression patterns. By capturing the co-expression of CXCL16/CXCR6 with other genes, scRNA-seq can position this signaling axis within broader transcriptional networks and identify cell states associated with CXCL16 responsiveness.
Single-cell proteomics and phosphoproteomics can reveal how CXCL16 stimulation triggers different signaling cascades across cell types, explaining differential functional responses. Mass cytometry (CyTOF) with CXCL16/CXCR6 antibodies allows simultaneous measurement of dozens of proteins across millions of cells, enabling high-dimensional phenotyping of expressing populations. Single-cell spatial transcriptomics maintains tissue organization information, revealing the spatial relationships between CXCL16-producing and CXCR6-expressing cells.
For functional studies, single-cell calcium imaging can capture real-time responses to CXCL16 across heterogeneous cell populations simultaneously. Combined with genetic lineage tracing using CXCR6-Cre driver lines, these technologies could track the fate and functional evolution of CXCR6-expressing cells during development and disease progression, revealing how CXCL16 shapes cellular identities and behaviors in complex biological contexts.
Several emerging research directions promise to transform our understanding of CXCL16 biology in the next decade. Structure-based drug design targeting the CXCL16-CXCR6 interface could yield selective modulators with therapeutic potential. Advances in cryo-electron microscopy may finally reveal the three-dimensional structure of CXCL16 bound to CXCR6, providing crucial insights into activation mechanisms and facilitating rational drug design.
Systems biology approaches integrating transcriptomics, proteomics, and metabolomics data from CXCL16/CXCR6-manipulated systems could reveal previously unrecognized connections to broader biological networks. Spatial multi-omics technologies maintaining tissue architecture information while providing molecular resolution will map CXCL16-CXCR6 signaling within complex tissue microenvironments, particularly valuable for understanding neuron-glia interactions and vascular functions.
Advanced genetic engineering using base editing and prime editing could generate precise mouse models with specific CXCL16 mutations corresponding to human variants or domain-specific modifications that selectively disrupt individual functions. Organoid systems derived from these mice would enable analysis of CXCL16 functions in more physiologically relevant three-dimensional contexts while reducing animal usage.
Finally, translational research connecting mouse findings to human disease through cross-species validation will be essential. Integration of mouse model data with human genetic association studies examining CXCL16/CXCR6 variants and disease susceptibility could identify high-priority therapeutic opportunities. These multidisciplinary approaches collectively promise to elevate CXCL16 from an interesting research target to a clinically relevant pathway with therapeutic potential across multiple disease contexts.