Fact-checked by Grok 2 weeks ago

CD69

CD69 is a disulfide-linked homodimeric type II of the superfamily, first identified in 1981 as an early marker on activated leukocytes, including T cells, B cells, natural killer () cells, and other hematopoietic cells. It is characterized by low expression in resting immune cells but rapid upregulation within hours of stimuli such as recognition or exposure. Encoded by a on , CD69 serves not only as a diagnostic indicator of early immune but also as a multifunctional regulator in various immune processes. Structurally, CD69 features an extracellular C-type lectin-like domain responsible for its carbohydrate-binding potential, a single transmembrane helix, and a short cytoplasmic lacking signaling motifs, with the homodimer stabilized by an interchain bridge between residues. The protein's molecular weight varies between 27 and 33 kDa due to differential N-linked , and its reveals a compact fold involving β-strands and α-helices that facilitate dimerization. This architecture enables CD69 to interact with other membrane proteins, such as the sphingosine-1-phosphate receptor 1 (S1PR1), through its , forming a cis-complex that promotes S1PR1 and degradation. Functionally, CD69 exerts immunoregulatory effects beyond marking, including the inhibition of egress from lymphoid organs by acting as an that promotes its Gi-dependent and , thereby disrupting S1P sensing and retaining activated cells at sites of during immune responses. It promotes (Treg) immunosuppressive activity and induces transforming growth factor-β (TGF-β) production in T cells, which downregulates excessive and , as evidenced in CD69-deficient models showing enhanced susceptibility to autoimmune diseases like collagen-induced . Additionally, CD69 is crucial for T and formation; it facilitates the homing and persistence of effector T-helper cells in the , enabling their into long-lived cells that support high-affinity production and in secondary responses. These roles highlight CD69's transition from a mere indicator to a pivotal gatekeeper in immune and response modulation.

Molecular Biology

Gene and Protein Structure

The CD69 gene, encoding a member of the superfamily, is located on the short arm of human at band p13.1 and on within the natural killer () gene complex, a genomic region harboring several immune-related genes. In both species, the gene spans approximately 8-9 kb and consists of five exons interrupted by four introns, with the coding sequence distributed across the exons to encode the full-length protein. The CD69 protein is a type II transmembrane that assembles into disulfide-linked homodimers on the surface, a configuration essential for its function as a receptor. The core polypeptide chain comprises 199 with a calculated of 22.6 per , but extensive post-translational results in an apparent molecular weight of 27-33 for individual subunits and up to 60 for the non-reduced dimer, as observed in analyses. Structurally, it features a short N-terminal cytoplasmic of about 40 rich in basic residues, a 21-amino-acid transmembrane , and an extracellular C-terminal region including a short stalk () domain followed by a C-type lectin-like (CTLD) of approximately 138 that mediates recognition and ligand interactions. The cytoplasmic lacks classical immunoreceptor tyrosine-based activation motifs but contains sequences that associate with intracellular signaling components, such as JAK3, to propagate signals upon receptor engagement. Post-translational modifications, particularly glycosylation, significantly influence CD69's maturation, stability, and membrane trafficking. The protein undergoes N-linked glycosylation at two extracellular sites: a typical motif (Asn-X-Thr) at position 166 and an atypical Asn-X-Cys motif at position 111, leading to heterogeneous glycoforms that form in the endoplasmic reticulum prior to Golgi processing. These N-glycans promote proper disulfide bond formation for dimerization and protect against degradation, while incomplete glycosylation can impair surface expression and trafficking. O-linked glycosylation, involving GalNAc attachment to serine or threonine residues in the stalk region, further contributes to glycan diversity and may modulate protein folding and stability, although its role is less dominant than N-glycosylation. CD69 exhibits strong evolutionary conservation across mammals, with orthologs identified in over 200 species including , , and , reflecting sequence identity of 50-90% in the CTLD and overall architecture. As part of the gene complex, it shares and structural homology with other C-type such as NKG2 family members in humans and Ly49 in mice, underscoring a common ancestral origin in the adaptive immune system's NK cell recognition machinery. This conservation highlights CD69's preserved role in early immune responses.

Expression Patterns

CD69 exhibits low constitutive expression in resting hematopoietic stem cells within the bone marrow niche, where it serves as a marker for quiescent progenitors, and on platelets, reflecting baseline surface presence without activation stimuli. This basal expression extends to certain tissue-resident immune cells, such as regulatory T cells (Tregs) in lymphoid organs and resident memory T cells (TRM) in non-lymphoid sites, distinguishing them from circulating counterparts. Upon immune activation, CD69 is rapidly upregulated within 2-3 hours in a broad array of leukocytes, including T cells (CD4+, CD8+, γδ T cells), B cells, natural killer (NK) cells, monocytes, and dendritic cells (DCs). This induction occurs through diverse stimuli, such as T cell receptor (TCR) engagement via anti-CD3 antibodies, cytokine signaling including interleukin-2 (IL-2) and transforming growth factor-β (TGF-β), or Toll-like receptor (TLR) ligation with ligands like lipopolysaccharide (LPS). Transcriptional activation begins as early as 30-60 minutes post-stimulation, peaking before declining after 4-6 hours, underscoring its role as an early and transient activation marker. Tissue-specific expression patterns highlight CD69's enrichment in lymphoid organs like the and , where it marks activated lymphocytes during immune responses, as well as in inflamed tissues such as those affected by or . In mucosal sites, including the gut and , CD69 is prominently expressed on TRM cells, promoting their retention and surveillance against pathogens; for instance, the majority of intestinal CD4+ and CD8+ TRM co-express CD69 alongside CD103. Similarly, skin-resident TRM upregulate CD69 shortly after tissue entry, facilitating long-term immunity. Expression of CD69 is tightly regulated at the transcriptional level by nuclear factor kappa B (), activator protein-1 (AP-1), and nuclear factor of activated T cells (NFAT) transcription factors, which bind promoter elements following activation signals. Post-transcriptional modulation occurs via microRNAs (miRNAs), such as miR-155, which enhances stability, and miR-20a or the miR-130/301 family, which inhibit expression by targeting CD69 mRNA. Quantitatively, CD69 surface expression is commonly assessed by , with detection thresholds typically set using isotype controls to identify positive cells above background (mean intensity >10^2-10^3 arbitrary units depending on antibody conjugates). This metric strongly correlates with activation states; for example, up to 50% of Tregs in secondary lymphoid organs constitutively express CD69 at moderate levels, while activated peripheral T cells can show >70% positivity within hours of stimulation.

Ligands and Signaling Pathways

Interacting Ligands

CD69, a C-type lectin-like receptor, engages several extracellular ligands primarily through its extracellular domain, influencing immune cell behavior in various microenvironments. The primary ligands include galectin-1 (Gal-1) and myosin light chains Myl9, Myl12a, and Myl12b (collectively Myl9/12). These interactions are calcium-independent, as the crystal structure of the CD69 C-type lectin-like domain (CTLD) reveals an absence of canonical calcium-binding residues, distinguishing it from classical C-type lectins. Gal-1 binds specifically to the CTLD of CD69 with high affinity, exhibiting a dissociation constant (Kd) of 96 nM as measured by (SPR) assays. This carbohydrate-dependent interaction was identified through pulldown experiments with on monocyte-derived dendritic cells and confirmed by (ELISA) and SPR, where binding was inhibited by , underscoring the role of . Gal-1 is ubiquitously expressed and secreted in the immune microenvironment by various cell types, including activated T cells and antigen-presenting cells, facilitating broad regulatory interactions with CD69-expressing leukocytes. In contrast, Myl9 and Myl12a/b function as tissue-specific ligands, predominantly localized to the and in inflamed sites, where they form net-like structures on the luminal surface of blood vessels. These non-muscle regulatory light chains bind directly to CD69 via electrostatic interactions with the N-terminal regions containing positively charged residues of Myl9 and Myl12a/b. Experimental validation includes co-immunoprecipitation and assays demonstrating specific association, with functional studies showing dependency for cellular retention in inflammatory contexts. Although specific Kd values for Myl9/12 binding remain unquantified, the interactions are robust and support CD69-mediated localization in inflammatory contexts. Additional ligands for CD69 include the S100A8/S100A9 complex and oxidized (ox-LDL), identified through and approaches. The S100A8/S100A9 heterodimer binds in a glycosylation-dependent manner with low nanomolar affinity, as assessed by with dissipation (QCM-D), and is secreted by activated myeloid cells in inflammatory settings. Ox-LDL engages the CTLD, promoting receptor , and is enriched in atherosclerotic plaques. Potential associations with or have been suggested in co-immunoprecipitation studies exploring complexes, though direct binding remains unconfirmed.

Downstream Signaling Mechanisms

Upon ligation by its interacting ligands, CD69 initiates intracellular signaling primarily through its short cytoplasmic tail, which associates with Janus kinase 3 (JAK3), leading to the and activation of signal transducer and activator of transcription 5 (STAT5). This JAK3/STAT5 pathway promotes (Treg) differentiation by upregulating expression and competes with to suppress Th17 cell development. In experimental models using CD69-deficient T cells, impaired STAT5 is observed, which can be restored by interleukin-2 supplementation, highlighting the pathway's role in maintaining immune balance. CD69 engagement also activates the mammalian target of rapamycin () pathway via interaction with the LAT1-CD98 amino acid transporter complex, enhancing nutrient uptake such as and driving metabolic reprogramming toward in activated lymphocytes. This mTOR activation stabilizes hypoxia-inducible factor 1-alpha (HIF1-α), further promoting glycolytic flux and effector T cell differentiation while inhibiting Treg function through degradation. Studies employing like rapamycin demonstrate reduced CD69-mediated metabolic shifts and altered T cell effector phenotypes, confirming the axis's centrality in cellular activation. In crosstalk with (TCR) signaling, CD69 amplifies downstream effectors such as extracellular signal-regulated kinase (ERK) and protein kinase C theta (PKCθ), facilitating enhanced T cell responses, while concurrently inhibiting (AMPK) to prioritize anabolic metabolism over catabolic processes. Phosphorylation assays in CD69-stimulated T cells reveal TGF-β-dependent suppression of ERK activation, underscoring context-specific modulation. Additionally, CD69 induces negative feedback through upregulation of suppressor of cytokine signaling 3 (SOCS3) via interactions with S100A8/S100A9, which limits excessive signaling and prevents prolonged inflammation. Experimental validation in CD69 knockout lymphocytes shows dysregulated JAK/STAT and pathways, leading to heightened Th17 responses and impaired Treg suppression, as evidenced by increased IL-17 production in autoimmune models like collagen-induced arthritis. These knockouts, combined with pathway-specific inhibitors, illustrate CD69's role as a metabolic gatekeeper in fine-tuning immune .

Immune Cell Functions

Activation Marker Role

CD69 serves as a well-established early activation marker for immune cells, particularly lymphocytes, and has been historically utilized in to detect rapid cellular responses to stimuli. Its expression is induced within 2-3 hours following (TCR) engagement or mitogenic stimulation, allowing for the identification of activated T cells, B cells, and natural killer () cells before the appearance of later markers such as CD25 ( α chain) or CD71 (). This temporal precedence makes CD69 a sensitive indicator in assays assessing immune responsiveness, with surface expression detectable as early as under 2 hours post- via cytometric analysis of peripheral or tissue samples. In clinical and research settings, CD69 upregulation has been employed to monitor T cell in conditions like allograft rejection and responses, providing a quantifiable metric of early immune engagement. Beyond its role as a marker, CD69 actively contributes to the activation process by promoting entry and inhibiting in . Upon , CD69 signaling enhances metabolic pathways, including activation and transport via the LAT1-CD98 complex, which supports the upregulation of cyclins necessary for progression from G0 to and subsequent . In developing thymocytes, CD69-positive double-positive cells exhibit increased expression, an anti-apoptotic protein that protects against during positive selection, thereby facilitating survival and expansion of activated clones. proliferation assays demonstrate that disruption of CD69 function, such as through blockade or genetic deficiency, impairs lymphocyte expansion by attenuating these metabolic and survival signals, leading to reduced numbers in response to antigenic stimulation. In NK cells, CD69 engagement similarly amplifies outcomes, enhancing cytotoxic activity against target cells and boosting interferon-γ (IFN-γ) production, which further amplifies innate immune responses. Unlike late markers like CD45RO, which persist on memory T cells to denote long-term , CD69 displays transient : its expression peaks within 4-24 hours post-stimulation and declines thereafter, reflecting its specificity to acute phases rather than sustained functional states. This ephemerality underscores CD69's utility in distinguishing immediate proliferative bursts from chronic immune memory.

T Cell Differentiation

CD69 plays a pivotal role in regulating T cell differentiation, particularly in promoting the development and maintenance of specific subsets such as regulatory T cells (Tregs) and tissue-resident memory T cells (TRM), while modulating the balance between proinflammatory and suppressive lineages. In the context of Treg differentiation, CD69 enhances expression through synergy with TGF-β signaling, which upregulates TGF-β1 production in CD69+ Tregs, and IL-2-mediated pathways that increase STAT5 phosphorylation for Treg stability and function. CD69+ Tregs exhibit superior suppressive capacity compared to CD69- counterparts, primarily via elevated IL-10 secretion driven by c-Maf and , enabling more effective inhibition of effector T cell and attenuation of inflammatory responses in models like dextran sulfate sodium-induced . CD69 is essential for the formation and retention of TRM cells in non-lymphoid s, where it modulates such as CD103 (αEβ7) to promote adhesion to epithelial cells via E-cadherin binding, thereby anchoring TRM in barrier sites. By sequestering 1 (S1PR1), CD69 inhibits egress signals, ensuring long-term tissue residency; this mechanism is critical for mucosal immunity, as CD69+ TRM in sites like the and female reproductive tract provide rapid protection against pathogens such as and . CD69 influences the Th17 versus Th1 balance by exerting an inhibitory effect on RORγt expression through S1PR1 antagonism, which limits /HIF-1α activation and signaling required for Th17 polarization, thereby reducing pathogenic Th17 responses in . In CD69-deficient models, exacerbated Th1 and Th17 differentiation leads to heightened inflammation in conditions like and , underscoring CD69's role in favoring regulatory over proinflammatory subsets. During memory T cell formation, CD69 sustains IL-7Rα expression on CD8+ memory precursors, supporting their survival and differentiation into long-lived memory populations, particularly in tissue-resident contexts where CD69+ CD103+ CD8+ cells show elevated CD127 (IL-7Rα) levels. In vivo studies using CD69-deficient mice reveal impaired Treg homeostasis, with reduced Treg stability and numbers leading to dysregulated immune suppression, as observed in models of myocardial infarction and intestinal inflammation. These mice also exhibit altered TRM populations, including marked reductions in skin CD8+ TRM and disruptions in gut mucosal TRM maintenance, highlighting CD69's necessity for proper subset development and tissue-specific immunity.

Lymphocyte Retention and Migration

CD69 plays a critical role in regulating trafficking by antagonizing the 1 (S1PR1), thereby inhibiting egress from lymphoid organs such as lymph nodes and the . Upon activation, CD69 is rapidly upregulated and directly binds to S1PR1, forming a complex that downregulates its surface expression and impairs S1PR1-mediated toward (S1P) gradients in lymphatic efferents. This interaction traps newly activated within lymphoid tissues, allowing time for maturation and before recirculation. Studies using co-expression assays in cell lines and primary have confirmed that CD69 specifically targets S1PR1, not related receptors like S1PR3, to enforce this retention mechanism. In addition to preventing egress, CD69 enhances lymphocyte retention in peripheral tissues by contributing to the tissue-resident T (TRM) , where it co-exists with such as αEβ7 (CD103) that mediate to epithelial s. In barrier sites like the gut and , CD69 expression on activated s supports prolonged residency, often alongside CD103, which binds E-cadherin on epithelial surfaces to anchor s against migratory cues. This dual mechanism—initial S1PR1 antagonism followed by integrin-mediated —ensures localized immune surveillance and rapid response in epithelial environments. Experimental evidence from models shows that CD69-deficient s exhibit reduced in these tissues, highlighting its role in stabilizing residency. CD69 also aids in resolving by limiting excessive recruitment through downregulation of receptors such as CCR7 and . As an early marker, CD69 suppresses the expression of these receptors on activated T cells, reducing their responsiveness to homeostatic like CCL19 (for CCR7) and (for ), which otherwise drive recirculation and infiltration. In murine models of intestinal , CD69-deficient + T cells displayed heightened levels, leading to increased migration and colonic accumulation, which exacerbated disease. This regulatory function dynamically balances retention during with controlled release post-maturation, preventing chronic . Evidence from CD69 knockout (KO) mice further underscores these roles, with assays revealing hyper-egress of lymphocytes from lymphoid organs and impaired tissue retention. In poly(I:C)-treated or virus-infected CD69 KO mice, lymphocytes showed accelerated S1P-directed migration and reduced sequestration in lymph nodes compared to wild-type controls, confirming CD69's necessity for spatiotemporal control of trafficking. Migration assays in vitro and adoptive transfer experiments in vivo demonstrated that CD69 KO cells exhibit enhanced chemotaxis toward multiple ligands, linking the phenotype directly to loss of retention signals. These models illustrate how CD69 induction upon activation creates a temporary "trap" for maturing lymphocytes, ensuring coordinated immune responses.

Clinical and Pathophysiological Roles

Involvement in Inflammatory Diseases

CD69 exhibits a in inflammatory diseases, serving as an early marker on immune cells while also contributing to regulatory mechanisms that suppress excessive inflammation. In the context of (IBD) and experimental models, CD69 expression on regulatory T cells (Tregs) enhances their immunosuppressive function, particularly through increased production of interleukin-10 (IL-10), which protects against intestinal inflammation. Studies in mice have shown that CD69 deficiency exacerbates dextran sodium sulfate (DSS)-induced , leading to heightened disease severity and immune dysregulation, as observed in post-2015 research. Human biopsy data from inflamed colonic tissues further demonstrate upregulated CD69 on infiltrating lymphocytes, underscoring its association with active inflammation in IBD patients. In (RA), CD69 is elevated on T cells, where its expression correlates directly with disease activity, positioning it as a potential for monitoring joint inflammation. Analysis of synovial samples from RA patients reveals significantly higher percentages of CD69-positive T cells compared to peripheral blood or healthy controls, reflecting ongoing T cell activation in the inflamed synovium. This upregulation contributes to the pro-inflammatory milieu but also highlights CD69's role in tissue-resident immune responses that may influence disease progression. CD69 also modulates cardiovascular inflammation, as evidenced in and post-myocardial (MI) settings. In a 2022 study, CD69 expression on Tregs limited infiltration into the heart and promoted Treg suppressive functions, thereby reducing immune-mediated damage after MI in both models and patients, where CD69 overexpression on Tregs was observed in peripheral blood post-event. This protective mechanism helps mitigate excessive inflammatory responses in the cardiac tissue. In and allergic conditions, CD69 regulates activation and Th2-driven responses in the airways; its absence leads to worsened allergen-induced eosinophilic inflammation and airway hyperresponsiveness, while expression on serves as a marker of activation in asthmatic tissues. Overall, CD69's dual functionality—promoting early immune activation to initiate responses while later enforcing suppression via Tregs and regulation—manifests across these conditions, with consistent upregulation in human biopsies from inflamed sites such as synovium, colon, and airways. This balance underscores CD69's potential as a therapeutic modulator in autoimmune and chronic inflammatory disorders, though its precise timing-dependent effects require further elucidation.

Applications in Cancer Immunotherapy

CD69 expression on (TILs) serves as a in various cancers, with high levels of CD69+ TILs generally associating with improved patient survival. In , elevated CD69+CD103+ + tissue-resident memory T (TRM) cells in tumors correlate with better outcomes in immunotherapy-naïve patients, reflecting enhanced antitumor immunity. Similarly, in non-small cell , increased CD69 expression on exhausted + TILs indicates a responsive immune state linked to favorable prognosis, while in models, CD69+ TILs predict reduced tumor progression and improved survival when associated with decreased T cell exhaustion. These associations highlight CD69's role in distinguishing exhausted yet potentially reinvigoratable T cells from terminally dysfunctional ones, providing prognostic value across solid tumors. In , CD69 upregulation following blockade, such as PD-1 inhibition, signals T cell reinvigoration and therapeutic response. Post-PD-1 blockade, CD69 expression rises on TILs, marking early activation and correlating with enhanced antitumor activity, as observed in and models where it predicts survival benefits. Combining anti-CD69 antibodies with PD-1 inhibitors further amplifies efficacy by alleviating exhaustion, leading to greater tumor regression in preclinical studies. This dynamic positions CD69 as an indicator of checkpoint therapy success, enabling monitoring of T cell responsiveness without invasive biopsies. CD69 also plays a key role in engineering chimeric antigen receptor (CAR) T cells for improved tumor retention, leveraging its function in promoting tissue residency. By inducing CD69 expression on CAR-T cells, therapies enhance their persistence within the tumor microenvironment, mimicking TRM cells to counteract egress and exhaustion; preclinical models show that CD69-mediated retention boosts local antitumor effects in solid tumors. This approach addresses a major limitation in CAR-T efficacy against solid malignancies, where poor infiltration and retention hinder outcomes. As a metabolic gatekeeper, CD69 supports TIL persistence in hypoxic tumor environments by modulating energy pathways. In , CD69 is directly upregulated as a HIF-1α target gene, inhibiting sphingosine-1-phosphate receptor 1 (S1P1)-mediated egress and promoting residency while restraining excessive to prevent metabolic burnout. This adaptation aids TIL survival and function in nutrient-scarce tumors, enhancing their longevity and effector potential during . Preclinical studies utilize CD69 as a pharmacodynamic marker to assess responses, particularly in anti-CTLA-4 regimens. In models, CD69 imaging via immuno-PET tracks early T cell activation post-CTLA-4/PD-1 blockade, correlating with prolonged survival. Preclinical extensions to anti-CTLA-4 combinations in other cancers suggest CD69 monitoring could optimize dosing and predict efficacy.

Emerging Therapeutic Targets

Recent preclinical studies have explored CD69 agonists to enhance , particularly through modulation of (Treg) function in autoimmune diseases. The interaction between CD69 and galectin-1 (Gal-1) plays a key role in suppressing Th17 cell differentiation and promoting , as CD69 binding to Gal-1 limits pathogenic Th17 responses while supporting Treg activity. Although specific Gal-1 mimetics targeting this pathway post-2020 remain in early patent stages without , strategies to activate CD69 signaling via Gal-1 analogs aim to boost Treg-mediated suppression in conditions like and . Antagonistic approaches targeting CD69-S1PR1 interactions offer potential for enhancing trafficking in cancer therapy. CD69 physically associates with S1PR1, promoting its and to retain activated lymphocytes in lymphoid tissues, thereby inhibiting egress to peripheral sites like tumors. with anti-CD69 monoclonal antibodies disrupts this complex, rapidly mobilizing hematopoietic stem/progenitor cells (HSPCs) and lymphocytes into circulation via restored S1PR1 function and activation. In preclinical models, this mobilization increases peripheral immune cell numbers without broad depletion. Monoclonal antibodies against CD69 have shown promise in preclinical models of inflammatory diseases, including (IBD) and -like conditions, by selectively reducing . In dextran sulfate sodium (DSS)-induced colitis models, anti-CD69 mAbs significantly ameliorated disease severity, decreased weight loss, and lowered pro-inflammatory levels (e.g., IFN-γ and IL-17) without causing systemic , as evidenced by preserved immune responses to unrelated antigens. Similar protective effects were observed in airway models relevant to comorbidities, where CD69 blockade attenuated Th17-driven responses. Although phase I clinical trials for anti-CD69 mAbs in IBD and are not yet reported, these findings support advancement to human studies for in mucosal . Gene therapy strategies involving CD69 overexpression in cells or Tregs represent an emerging avenue for desensitization. Overexpression of CD69 in Foxp3+ Tregs enhances their immunosuppressive capacity by promoting IL-10 production and attenuating in adoptive transfer models, suggesting potential for sustained induction. In allergic contexts, CD69 intrinsically limits type 2 immune responses and activation; thus, engineered overexpression in hematopoietic cells could facilitate long-term desensitization to allergens by reinforcing regulatory pathways during . Preclinical data indicate that CD69+ Tregs restore and prevent inflammation in models, positioning this approach for combination with allergen-specific therapies. Recent preclinical advances as of 2025 include development of a novel CD69-targeted PET tracer for monitoring T cell activation in immunotherapy and assessment of anti-CD69 antibody therapy, alone or combined with anti-PD-1, in murine glioblastoma models to improve anti-tumor immunity. Despite these advances, therapeutic targeting of CD69 faces challenges related to specificity and off-target effects due to its broad expression on activated immune cells. The multifaceted signaling pathways of CD69, including associations with S1PR1, LAT1-CD98, and PD-1 induction, complicate selective modulation without disrupting homeostasis. A 2023 review highlights the need for inhibitors of downstream signaling (e.g., via myosin light chains or S100 proteins) to achieve precise control. Future directions emphasize combinations with checkpoint inhibitors like anti-PD-1 to enhance anti-tumor immunity while addressing tissue residency, alongside improved delivery systems for gene therapies to minimize immunogenicity.

References

  1. [1]
    CD69 is a Promising Immunotherapy and Prognosis Prediction ... - NIH
    Jan 9, 2024 · Discovered in 1981, CD69 is a disulfide-linked homodimer protein ... Induction of tumor NK-cell immunity by anti-CD69 antibody therapy.Missing: history | Show results with:history
  2. [2]
    CD69 - an overview | ScienceDirect Topics
    CD69 is a type II C-lectin membrane receptor with a scarce expression in resting lymphocytes that is rapidly induced upon cell activation.
  3. [3]
  4. [4]
    Transmembrane protein CD69 acts as an S1PR1 agonist - eLife
    Apr 11, 2023 · CD69 is an endogenous negative regulator of lymphocyte egress that interacts with S1PR1 in cis to facilitate internalization and degradation of the receptor.
  5. [5]
  6. [6]
    Type II membrane protein CD69 regulates the formation of resting T ...
    Thus, CD69 is critical for the generation and maintenance of professional memory Th lymphocytes, which can efficiently help humoral immunity in the late phase.
  7. [7]
    Regional sublocalization of the human CD69 gene to chromosome ...
    Here we report the regional sublocalization of the human CD69 gene to chromosome bands 12p12.3-p13.2, suggesting that CD69 belongs to one linkage group together ...Missing: 12q13 | Show results with:12q13
  8. [8]
    The mouse CD69 gene. Structure, expression, and ... - PubMed - NIH
    Chromosomal mapping placed the mouse CD69 gene on the long arm of chromosome 6 near the NK gene complex that contains the related NKR-P1 and Ly-49 gene families ...Missing: 12q13 | Show results with:12q13
  9. [9]
    a C-type lectin receptor evolutionarily related with the gene families ...
    We have herein studied the genomic structure of the human gene encoding CD69. The coding sequence is divided into five exons separated by four introns. The ...Missing: organization | Show results with:organization
  10. [10]
    Phenotypic and functional characteristics of hematopoietic cell ...
    The CD69-mouse genomic DNA consists of 5 exons and 4 introns and is approximately 9 kb in length.7 To disrupt CD69-gene expression by homologous recombination ...
  11. [11]
    The high-resolution structure of the extracellular domain of human ...
    Human CD69 is a disulfide-linked homodimer with subunits of molecular mass 28 or 32 kDa resulting from different glycosylation at two possible extracellular N- ...
  12. [12]
    Early activation antigen CD69 - Homo sapiens (Human) - UniProt
    Transmembrane protein expressed mainly on T-cells resident in mucosa that plays an essential role in immune cell homeostasis.Missing: intron | Show results with:intron
  13. [13]
    CD69 Gene - GeneCards | CD69 Protein | CD69 Antibody
    CD69 (CD69 Molecule) is a Protein Coding gene. Diseases associated with CD69 include Coccidioidomycosis and Eosinophilic Pneumonia.Missing: organization | Show results with:organization
  14. [14]
    CD69 Monoclonal Antibody (FN50), PE (MA1-10276)
    6–10 day delivery... 27.. Learn ... CD69 (AIM, Active Inducer Molecule) is a gp28/34 disulfide bonded homodimer with a molecular weight of 60 kDa under non-reducing conditions.
  15. [15]
  16. [16]
    Crystal Structure of the C-type Lectin-like Domain from the Human ...
    Human CD69 consists of a 40-residue intracellular domain, a 21-residue transmembrane region, and an extracellular portion that comprises a 20-residue neck and ...
  17. [17]
    Unraveling CD69 signaling pathways, ligands and laterally ... - NIH
    CD69 is a glycosylated homodimeric receptor formed by two disulphide-linked chains of 28 and 32 KDa (Testi et al., 1989[77]; López-Cabrera et al., 1993[40]).
  18. [18]
    Multiple dimeric forms of human CD69 result from differential ...
    Sep 12, 1997 · In the current report we show that human CD69 glycoforms are generated before the egress of CD69 proteins from the endoplasmic reticulum to the ...Missing: O- | Show results with:O-
  19. [19]
    Multiple Dimeric Forms of Human CD69 Result from Differential ...
    Multiple dimeric forms of human CD69 result from the variable addition of N-glycans to atypical and typical glycosylation motifs within the CD69 extracellular ...
  20. [20]
    Distinct but dispensable N-glycosylation of human CD69 proteins
    Human CD69 is uniquely glycosylated at typical (Asn-X-Ser/Thr) and atypical (Asn-X-Cys) motifs, which represents the molecular basis for the formation of CD69Missing: apparent weight
  21. [21]
    Gene: CD69 (ENSG00000110848) - Summary - Ensembl
    Chromosome 12: 9,752,486-9,760,901 reverse strand. This gene has 4 transcripts (splice variants), 217 orthologues and 23 paralogues. .Missing: organization | Show results with:organization
  22. [22]
    Evolution of the C-Type Lectin-Like Receptor Genes of the DECTIN ...
    The human C-type lectin-like receptors encoded in the DECTIN-1 cluster within the NK gene complex contain prominent receptors with pattern recognition function, ...
  23. [23]
    CD69: from activation marker to metabolic gatekeeper - PMC
    The cytoplasmic tail of CD69 triggers intracellular signals through JAK3, which then phosphorylates and activates STAT5 (Fig. 1) [23].Missing: motifs | Show results with:motifs
  24. [24]
    Tissue resident memory T cells in mice and humans - PubMed Central
    CD69, originally defined as an early T cell activation marker, is expressed by a large proportion of tissue memory CD4+ and CD8+ T cells in mice and humans and ...
  25. [25]
    Optimized flow cytometric protocol for the detection of functional ...
    CD69 is an early-activated marker expressed by CD4+ and CD8+ T cells. Its rapid expression (<2 h post-activation) on a broad range of immune cells (T cells, B ...
  26. [26]
    CD69 gene is differentially regulated in T and B cells by ... - NIH
    The CD69 gene is located in the natural killer complex (NKC) on mouse chromosome 6 and human chromosome 12. This complex includes a variety of genes encoding C ...
  27. [27]
    CD69 Is the Crucial Regulator of Intestinal Inflammation
    CD69 regulates intestinal inflammation by directing immune responses towards oral tolerance and regulatory responses, limiting inflammation in the gut.
  28. [28]
    An Overview of Tissue-Resident Memory T Cells in the Intestine
    Lectin CD69, a marker for early T cell activation, is reexpressed in the intestinal TRM, and it can be used to distinguish TRM from their circulating ...
  29. [29]
    Pathophysiology of Skin Resident Memory T Cells - PMC
    CD69 is upregulated shortly after memory T cells reaching the skin and CD69 expression is critical for early T cell retention rather than recruitment of T cell ...
  30. [30]
    miR-20a Inhibits TCR-Mediated Signaling and Cytokine Production ...
    Apr 17, 2015 · When overexpressed in human naïve CD4+ T-helper cells, miR-20a inhibits TCR-mediated signaling, CD69 expression, and decreases cytokine ...
  31. [31]
    Non-coding RNAs in CD8 T cell biology - PMC - PubMed Central - NIH
    Apr 1, 2021 · It has been reported that downregulation of CD69 is controlled by the miR-130/301 family of miRNAs that are gradually induced upon TCR-signaling ...
  32. [32]
    A Whole-Blood Assay for Qualitative and Semiquantitative ... - NIH
    The assay uses flow cytometry to measure CD69 surface expression on CD4 and CD8 T-lymphocytes, assessing T-lymphocyte activation using whole-blood samples.
  33. [33]
    Activation-Induced Marker Assay to Identify and Isolate HCV ...
    Oct 17, 2024 · CD69 has been used to identify antigen-specific CD8 T cells upon TCR stimulation, with peak expression at 18 h [2]. However, expression of CD69 ...Missing: thresholds | Show results with:thresholds
  34. [34]
    The Leukocyte Activation Receptor CD69 Controls T Cell ... - NIH
    CD69 is involved in immune cell homeostasis, regulating the T cell-mediated immune response through the control of Th17 cell differentiation.Missing: thresholds | Show results with:thresholds
  35. [35]
    Myosin light chains 9 and 12 are functional ligands for CD69 that ...
    We herein identified myosin light chain (Myl) 9, Myl12a, and Myl12b (Myl9/12) as new functional ligands for CD69 in inflamed lungs. Upon airway inflammation, ...Missing: text | Show results with:text
  36. [36]
    CD69 enhances immunosuppressive function of regulatory T-cells ...
    Sep 5, 2018 · Collective findings show that CD69 functions as a molecule involved in the regulation of immune response rather than a simple activation marker.
  37. [37]
    CD69: from activation marker to metabolic gatekeeper - PubMed
    CD69 is a membrane-bound, type II C-lectin receptor. It is a classical ... Signal Transduction; T-Lymphocyte Subsets / immunology*; T-Lymphocyte Subsets ...
  38. [38]
    Increased expression of the lymphocyte early activation marker ...
    Increased expression of the lymphocyte early activation marker CD69 in peripheral blood correlates with histologic evidence of cardiac allograft rejection.
  39. [39]
    The expression of Bcl-2 family proteins (Bcl-2, Bcl-x, Bax, Bak and ...
    Among DP thymocytes, cells co-expressing CD69 up-regulated Bcl-2, suggesting that the role of Bcl-2 is promoting survival of positively selected DP cells.
  40. [40]
    Differential effect of CD69 targeting on bystander and antigen ...
    Jul 1, 2012 · The anti-CD69 2.2 mAb was also able to induce T cell proliferation in in vitro cultures of whole LNs and spleen cells. In the case of CD8+ T ...
  41. [41]
    CD69 is a stimulatory receptor for natural killer cell and its cytotoxic ...
    The results show that CD69-mediated NK cytotoxicity can be abrogated by CD94 stimulation in NK cells expressing the CD94 inhibitory form of the receptor.
  42. [42]
    Antigen specific T cell analysis reveals active immune responses to ...
    While activation markers like CD69 and CD25 can be used to define T cell activation status, their upregulation is relatively transient [31, 32]. Expression of ...
  43. [43]
    Article Human intestinal tissue-resident memory T cells comprise ...
    Jan 19, 2021 · CD103+ CD8+ T cells expressed more CD161 and CD127 (IL-7R) compared with the CD69− or CD69+CD103− cells, consistent with the transcriptomic data ...
  44. [44]
    CD69 expression protects from immune damage after MI - JCI
    Sep 6, 2022 · CD69+ Tregs induce apoptosis and reduce IL-17A production in γδT cells in a CD39-dependent manner in mice. Previous studies provide evidence for ...
  45. [45]
    CD69: from activation marker to metabolic gatekeeper - Cibrián - 2017
    May 5, 2017 · ... cytoplasmic tail (Fig. 1). The CD69 gene is located in the natural killer (NK) gene cluster, chromosome 6 in mouse and 12 in human 1, 2.
  46. [46]
  47. [47]
  48. [48]
    CD69 Is the Crucial Regulator of Intestinal Inflammation: A New ...
    Feb 22, 2015 · To summarize we believe that CD69 regulates TGF-β production by T cells and may serve as a regulatory molecule in the immune system. To further ...
  49. [49]
    CD69 on synovial T cells in rheumatoid arthritis correlates ... - PubMed
    CD69 on synovial T cells in rheumatoid arthritis correlates with disease activity. Br J Rheumatol. 1996 Apr;35(4):397. doi: 10.1093/rheumatology/35.4.397 ...
  50. [50]
    Expression of CD69 antigen on synovial fluid T cells in patients with ...
    Results: The percentage of T cells bearing CD69 was significantly increased in synovial fluid from patients with RA (30.3 (13)%) and other chronic synovitis (18 ...Missing: activity | Show results with:activity
  51. [51]
    Expression of CD69 antigen on synovial fluid T cells in patients with ...
    RESULTS--The percentage of T cells bearing CD69 was significantly increased in synovial fluid from patients with RA (30.3 (13)%) and other chronic synovitis (18 ...Missing: elevated activity
  52. [52]
    CD69 expression on regulatory T cells protects from immune ... - NIH
    Nov 1, 2022 · CD69 expression drives CD39 expression in myocardial Treg and T effector cells, although CD69+ Tregs exhibited the highest CD39 levels ( ...
  53. [53]
    CD69 expression on regulatory T cells protects from immune ...
    In this study, a broad analysis of immune markers in 283 patients revealed significant CD69 overexpression on Tregs after MI. Our results in ...
  54. [54]
    Crucial role for CD69 in allergic inflammatory responses: CD69 ...
    Jun 28, 2017 · We recently elucidated a novel mechanism, in which the interaction between CD69 and its ligands, myosin light chain 9, 12a and 12b (Myl9/12) ...Missing: Myl12a | Show results with:Myl12a
  55. [55]
    CD69 controls the pathogenesis of allergic airway inflammation
    Dec 15, 2009 · CD69 plays a crucial role in the pathogenesis of allergen-induced eosinophilic airway inflammation and hyperresponsiveness.Missing: regulates | Show results with:regulates
  56. [56]
    Immuno-PET Imaging of CD69 Visualizes T-Cell Activation and ...
    CD69 is Upregulated Upon T-Cell Activation In Vitro. To corroborate our hypothesis that CD69 is an ideal imaging agent for monitoring ICI response by virtue ...
  57. [57]
    Full article: CD69 is a Promising Immunotherapy and Prognosis ...
    The binding of Gal-1, S100A8/S100A9 complexes, and ox-LDL to CD69 may be involved in the negative regulation of immune response, whereas the binding of Myl9/12 ...
  58. [58]
    CD69 is a direct HIF-1α target gene in hypoxia as a mechanism ...
    CD69 is a direct HIF-1α target gene in hypoxia, with its expression enhanced on tumor-infiltrating T lymphocytes, and is upregulated in hypoxia.Results · Material And Methods · Mice And Cell Lines
  59. [59]
    Enhanced Susceptibility of Galectin-1 Deficient Mice to Experimental ...
    Jun 27, 2021 · We found that galectin-1 deficient mice (Lgals1 −/− mice) displayed a more severe intestinal inflammation, characterized by significantly elevated clinical ...Missing: mimetics | Show results with:mimetics
  60. [60]
    Transmembrane protein CD69 acts as an S1PR1 agonist - bioRxiv
    Feb 15, 2023 · Thus, our structural findings along with functional analyses demonstrate that CD69 acts in cis as a protein agonist of S1PR1, thereby promoting ...<|control11|><|separator|>
  61. [61]
    Anti-CD69 therapy induces rapid mobilization and high proliferation ...
    Blockade of CD69 with monoclonal antibodies induced a rapid mobilization of large numbers of BM leukocytes, which is inhibited by using the agonist of S1P1 ...Missing: disruptors | Show results with:disruptors
  62. [62]
    Anti-CD69 therapy induces rapid mobilization and high ... - Sabio lab
    Feb 27, 2018 · CD69 regulates lymphocyte egress from the thymus and lymph nodes through cis-interactions and the downregulation of surface sphingosine-1 ...Missing: antagonists disruptors
  63. [63]
    Crucial Role for CD69 in the Pathogenesis of Dextran Sulphate ...
    ... CD69 KO mice restored the induction of colitis. The administration of an anti-CD69 antibody also inhibited the induction of the DSS-induced colitis. These ...Missing: preclinical | Show results with:preclinical
  64. [64]
    The leukocyte activation antigen CD69 limits allergic asthma and ...
    CD69 regulates the immune response to antigen-induced airway inflammation by limiting TH2 and TH17 responses in the lungs. •. In hapten-induced CHS, loss of ...
  65. [65]
    Maintenance of immune tolerance by Foxp3 + regulatory T cells ...
    L.R. Shiow et al. CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature. (2006). M. Lopez-Cabrera et ...
  66. [66]
    Unraveling CD69 signaling pathways, ligands and laterally ...
    Mar 16, 2023 · This review provides a perspective on the molecular pathways, ligands and cellular functions known to be regulated by CD69.