CD79A is a 226-amino acid protein with distinct domains:
Extracellular immunoglobulin (Ig) domain: Facilitates heterodimerization with CD79B via disulfide bonds .
Transmembrane region: Anchors the BCR complex to the cell membrane .
Cytoplasmic immunoreceptor tyrosine-based activation motif (ITAM): Contains dual phosphotyrosine residues (Tyr188/Tyr199 in humans) critical for signal transduction .
Two splice variants exist:
| Isoform | Length | Key Feature |
|---|---|---|
| Isoform 1 | 226 aa | Full Ig domain |
| Isoform 2 | 196 aa | Truncated Ig domain |
CD79A and CD79B form a 1:1 heterodimer, which non-covalently associates with membrane-bound immunoglobulins (IgM/IgD) to complete BCR assembly .
CD79A is indispensable for:
BCR surface expression: Loss of CD79A blocks B-cell development at the pro-B to pre-B transition .
Signal transduction: ITAM phosphorylation recruits SYK kinase, initiating downstream pathways (PI3K-AKT, MAPK) .
Tonic signaling: Maintains basal B-cell survival via low-level BCR activity .
Key functional studies:
CD79A is a pan-B-cell marker used in immunohistochemistry due to its:
Ubiquitous expression across B-cell lineages (from pro-B cells to plasma cells) .
Retention in >95% of B-cell neoplasms, including lymphomas and myelomas .
| Condition | Mechanism | Clinical Impact |
|---|---|---|
| Agammaglobulinemia-3 | Homozygous CD79A mutations causing BCR deficiency | Autosomal recessive immune deficiency |
| B-cell precursor ALL (BCP-ALL) | High CD79A expression correlates with CNS relapse | 8-fold increased CNS infiltration risk |
| Autoimmune disorders | CD79A ITAM mutations alter BCR signaling thresholds | Potential therapeutic target |
Recent preclinical studies highlight CD79A’s therapeutic potential:
Anti-CD79A antibodies (Fc-inert):
CD79A knockdown in BCP-ALL xenografts:
Data from The Human Protein Atlas :
| Tissue | Expression Level | Localization |
|---|---|---|
| Lymph node | High | Germinal centers |
| Bone marrow | Moderate | Immature B-cells |
| Spleen | High | Marginal zone |
CD79A interacts with:
CNS Leukemia: CD79A overexpression promotes leptomeningeal infiltration in pediatric BCP-ALL via pre-BCR signaling .
B-Cell Anergy: Transient CD79A modulation could treat autoimmunity without broad immunosuppression .
Lymphoma Therapy: CD79A-directed CAR-T cells show preclinical efficacy in CD79A+ malignancies .
CD79a Molecule, Membrane-Bound Immunoglobulin-Associated Protein, CD79A Antigen (Immunoglobulin-Associated Alpha), CD79a Molecule, Immunoglobulin-Associated Alpha, Surface IgM-Associated Protein, MB-1 Membrane Glycoprotein, Ig-Alpha, IGA, B-Cell Antigen Receptor Complex-Associated Protein Alpha Chain, CD79a Antigen, MB-1, MB1.
Sf9, Insect cells.
LWMHKVPASL MVSLGEDAHF QCPHNSSNNA NVTWWRVLHG NYTWPPEFLG PGEDPNGTLI IQNVNKSHGG IYVCRVQEGN ESYQQSCGTY LRVRQPPPRP FLDMGEGTKN RLEHHHHHH.
Human CD79A (also known as Mb-1, Ig alpha, and B cell antigen receptor complex-associated protein alpha-chain) is a 30-40 kDa type I transmembrane glycoprotein belonging to the immunoglobulin superfamily. The mature protein contains an extracellular region (amino acids 33-143) with one C2-type Ig-like domain (amino acids 33-116), and an immunoreceptor tyrosine-based activation motif (ITAM)-containing cytoplasmic domain (amino acids 177-205) .
CD79A forms a covalent heterodimer with CD79B through disulfide bonding. These heterodimers interact non-covalently with membrane immunoglobulin (Ig), forming the B cell antigen receptor (BCR) complex . Within this complex, membrane Ig functions to detect antigen, while the CD79A/B heterodimer initiates intracellular signaling through their cytoplasmic ITAM domains . This signaling is critical for B cell development, survival, and antigen-induced activation .
CD79A plays an essential role in B cell differentiation, particularly at the pre-B cell stage . In the pre-B cell receptor (pre-BCR), CD79A participates in signaling that regulates surface expression of IL-7R, an important receptor for B cell development .
The functional importance of CD79A is demonstrated in knockout models. Studies show that BCR-ABL1-transformed pro-B cells lacking CD79A exhibit significant delays in leukemic engraftment in the spleen, bone marrow, and central nervous system (CNS) when injected into mice . Animals receiving CD79A-knockout cells showed a median survival prolongation of 66 days compared to control cells (95 days versus 29 days) . These findings suggest that CD79A is indispensable for leukemia development, particularly for CNS involvement.
Importantly, the role of CD79A extends beyond its direct signaling function, as downstream pathways including ZAP70, PI3K, and MAPK, which act downstream of the pre-BCR, have all been shown to be directly involved in CNS involvement in leukemia .
Several methodological approaches have proven effective for CD79A research:
Flow Cytometry: Detection of CD79A in human blood monocytes and peripheral blood mononuclear cells (PBMCs) can be accomplished using monoclonal antibodies. Typically, cells are co-stained with CD19 to identify the B cell population, followed by CD79A staining. Control antibodies are essential for setting appropriate quadrant markers .
Recombinant Protein Studies: Recombinant human CD79A protein (such as the extracellular domain Leu33-Arg143 with a C-terminal 6-His tag) can be expressed and purified for functional studies . The protein can be analyzed using SDS-PAGE under reducing and non-reducing conditions, typically showing bands at 38-43 kDa .
Genetic Modification: CRISPR/Cas9 techniques have been used to generate CD79A knockout cells. In one study, CRISPR-Cas9-mediated CD79a gene editing was performed in mouse fetal liver-derived pro-B-cells, which were then cultured with IL-7 and subsequently transformed with BCR-ABL1 .
Humanized Mouse Models: To overcome the lack of cross-reactivity between human therapeutic antibodies and mouse CD79A, knockin mice have been generated in which the extracellular Ig-like domains of CD79A and CD79B are replaced with human equivalents .
A significant correlation has been identified between CD79A expression and central nervous system (CNS) infiltration in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Analysis of patient cohorts has revealed:
CD79A levels significantly correlate with ZAP70 and IL7R mRNA levels in BCP-ALL .
Patients diagnosed as CNS-positive show significantly higher levels of CD79A than CNS-negative patients (median CD79a expression: 5.183 ± 0.396 in CNS− vs. 7.537 ± 1.278 in CNS+) .
Multivariate logistic regression analysis demonstrates that CD79A expression above the 75th percentile is associated with approximately 8-fold increased risk for CNS-positivity compared to the lower quartile (odds ratio = 7.873, 95% CI [1.338, 46.312], p = 0.022) .
Even CD79A expression levels above the 25th percentile show a significant 7-fold increased risk for CNS-positivity (odds ratio = 7.0, 95% CI [1.4, 33.9], p = 0.016) .
In an independent cohort from the TARGET phase 1 data set, increased expression of CD79A (z-score ≥ 1.2) was associated with significantly reduced long-term probability rates for isolated-CNS-relapse-free survival .
These findings suggest that CD79A could potentially serve as a surrogate marker for CNS involvement in BCP-ALL and could be important for understanding disease progression and relapse.
Anti-CD79A antibodies represent a promising therapeutic approach for autoimmune diseases and B cell malignancies . Key research findings include:
Non-depleting Antibody Approach: Anti-human CD79A antibodies that lack Fc region effector function (preventing antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity) do not cause significant B cell depletion but instead induce a transient form of B cell anergy . This mechanism differs from anti-CD20 therapies, which act by depleting B cells .
Induced Changes in B Cells: Treatment with these antibodies leads to:
Efficacy in Autoimmune Models: Anti-human CD79A treatment has shown efficacy in preventing disease development in multiple mouse models of autoimmunity, including experimental autoimmune encephalomyelitis (EAE) and pristane-induced production of anti-chromatin autoantibodies . In the EAE model, prophylactic treatment significantly reduced disease severity and led to complete recovery .
In vitro Effects on Plasma Cells: Evidence suggests that anti-human CD79A treatment may inhibit antibody secretion by terminally differentiated plasmablasts and plasma cells in vitro, extending its potential applications .
Due to limited cross-reactivity between human therapeutic antibodies and mouse CD79A (only 57% amino acid sequence identity in the extracellular domain), specialized animal models have been developed :
Humanized CD79 Knockin Mice: These mice have been generated by replacing the extracellular Ig-like domains of mouse CD79A and CD79B with human equivalents . The constructs are assembled using bacterial artificial chromosome (BAC) recombineering techniques including Red recombination and cleavage with homing endonuclease I-SceI .
Chimeric CD79A Design: For chimeric CD79A (cCD79A), amino acids 29-160 of mouse CD79A are replaced by amino acids 34-166 of human CD79A . This approach maintains the transmembrane and cytoplasmic domains (which have higher homology: 89% in CD79A) while introducing the human extracellular domain .
Functional Interchangeability: Research has demonstrated that human and mouse CD79 extracellular domains are functionally interchangeable, making these models suitable for preclinical testing of human-targeted therapies .
CD79A Knockout Models: CRISPR/Cas9 techniques have been used to generate CD79A knockout cells for functional studies and in vivo experiments .
Several technical challenges exist in CD79A research:
Protein Stability: Recombinant CD79A protein stability can be challenging. To address this, carrier proteins like Bovine Serum Albumin (BSA) are often added to enhance stability, increase shelf-life, and allow storage at more dilute concentrations . For applications where BSA might interfere, carrier-free versions are recommended .
Reconstitution and Storage: Proper reconstitution and storage protocols are crucial. Typically, lyophilized CD79A protein should be reconstituted at 250 μg/mL in PBS, and repeated freeze-thaw cycles should be avoided .
Cross-reactivity Issues: The limited homology between human and mouse CD79A extracellular domains (57%) complicates preclinical testing of human-targeted therapies in mouse models . This necessitates the generation of specialized knockin mice expressing human CD79A extracellular domains .
Detection Methods: For flow cytometry detection, appropriate control antibodies are essential for setting quadrant markers and avoiding false positives . Co-staining with CD19 helps identify the B cell population more accurately .
CD79A signaling operates through distinct mechanisms in normal and diseased states:
Normal B Cells: CD79A and CD79B contain cytoplasmic immunoreceptor tyrosine-based activation motifs (ITAMs) that mediate intracellular propagation of BCR signals critical for B cell development, survival, and antigen-induced activation . In normal B cells, these signals are tightly regulated.
Malignant B Cells: In malignancies like BCR-ABL+ acute lymphoblastic leukemia, even subgroups considered preBCR negative (lacking an assembled preBCR complex on the surface) show dependence on CD79A signaling . This suggests that the signaling molecules of the preBCR are indispensable for leukemia development and CNS involvement .
Downstream Pathways: ZAP70, PI3K, and MAPK pathways, which act downstream of CD79A, have been shown to be directly involved in CNS involvement in leukemia . This indicates that CD79A signaling extends beyond its immediate effects and influences multiple cellular processes.
Therapeutic Modulation: Anti-CD79A antibodies can act as "reverse agonists," inducing BCR desensitization without depleting B cells . This leads to a state of polyclonal B cell anergy that is lost upon decay of the antibody in vivo .
Several promising research directions are emerging:
CD79A as a Biomarker: The association between CD79A expression and CNS infiltration in BCP-ALL suggests its potential use as a prognostic biomarker . Further research could refine its application in risk stratification and treatment planning.
Combination Therapies: Exploring combinations of anti-CD79A therapies with other targeted approaches might enhance efficacy in both autoimmune diseases and B cell malignancies.
Selective Targeting of Pathogenic B Cells: Developing approaches that can selectively target pathogenic B cells while sparing protective ones could improve the therapeutic index.
Effects on Plasma Cells: Further investigation into the effects of anti-CD79A treatment on antibody secretion by terminally differentiated plasmablasts and plasma cells might extend its applications to antibody-mediated diseases .
Structure-based Drug Design: Detailed structural understanding of CD79A-CD79B-Ig interactions could facilitate the design of small molecule inhibitors or more specific antibodies targeting functional epitopes.
CD79A is a transmembrane protein that forms a heterodimer with CD79B (Ig-beta). This heterodimer is non-covalently associated with membrane-bound immunoglobulins (IgM, IgD, IgG, etc.), forming the BCR complex . The BCR complex is responsible for recognizing specific antigens and initiating B-cell activation and signaling .
The cytoplasmic domain of CD79A contains immunoreceptor tyrosine-based activation motifs (ITAMs), which are critical for signal transduction. Upon antigen binding to the BCR, the ITAMs are phosphorylated, leading to the recruitment and activation of downstream signaling molecules such as Lyn and Syk kinases . This signaling cascade ultimately results in B-cell activation, proliferation, and differentiation .
CD79A expression begins at the pre-B cell stage and continues through to the plasma cell stage . It is found in the majority of B-cell lineage cells, including precursor B cells, mature B cells, and some plasma cells . CD79A is not present in myeloid or T-cell lineages .
The presence of CD79A is essential for the proper assembly and surface expression of the BCR complex. It also plays a role in the selection and maturation of B cells in the bone marrow . Mutations or deficiencies in CD79A can lead to immunodeficiencies, such as agammaglobulinemia, where B cells are unable to produce sufficient antibodies .
Recombinant CD79A is produced using various expression systems, such as HEK293 cells, to ensure proper folding and post-translational modifications . The recombinant protein is often tagged with a His-tag for purification purposes and is used in research to study B-cell signaling and function .
Recombinant CD79A is also utilized in the development of therapeutic antibodies and chimeric antigen receptor (CAR) T-cell therapies targeting B-cell malignancies and autoimmune diseases . These therapies aim to modulate B-cell activity by targeting the CD79A component of the BCR complex .
CD79A is a valuable marker for diagnosing B-cell lymphomas and leukemias . Its expression is often assessed in immunohistochemistry and flow cytometry to identify B-cell lineage cells in various hematological malignancies . Additionally, CD79A-targeted therapies are being explored for their potential to treat B-cell-related disorders .