RAC2 Antibody is a specialized immunological reagent designed to detect and study Ras-related C3 botulinum toxin substrate 2 (RAC2), a small Rho-family GTPase critical for actin cytoskeleton remodeling, intracellular signaling, and immune cell functions . These antibodies enable researchers to investigate RAC2's role in hematopoiesis, neutrophil migration, T/B-cell development, and pathological conditions such as immunodeficiencies and cancers .
RAC2 antibodies are validated for multiple experimental techniques:
Neutrophil Defects: RAC2 mutations impair chemotaxis, superoxide production, and macropinosome formation, linked to leukocyte adhesion deficiency (LAD)-like symptoms .
B-Cell Dysfunction: RAC2-deficient mice show reduced IgM/IgA levels, impaired calcium flux, and defective plasma cell differentiation .
Inflammasome Activation: Gain-of-function RAC2 mutations (e.g., A59S, E62K) trigger NLRP3 inflammasome activity, correlating with autoinflammatory syndromes .
Clear Cell Renal Cell Carcinoma (ccRCC): Elevated RAC2 expression correlates with poor survival (HR = 1.67, P = 0.006) and advanced tumor stages .
Immunodeficiency Syndromes:
Therapeutic Targeting: Small-molecule inhibitors of RAC2 (e.g., NSC23766) are under investigation for inflammatory diseases and cancers .
RAC2 is a member of the Rho family of small GTPases, specifically a 21.4 kilodalton protein that functions as a molecular switch in immune cells. It is known by several alternative names including EN-7, Gx, HSPC022, p21-Rac2, and ras-related C3 botulinum toxin substrate 2 . RAC2 is predominantly expressed in hematopoietic cells and plays crucial roles in B cell development, adhesion, and immunological synapse formation .
Unlike its more ubiquitous family member RAC1, RAC2 has specialized functions in immune cells, with studies showing that RAC2 is more important than RAC1 for B cell development and function . RAC2's significance extends to neutrophil activation, T cell development, and antigen-specific antibody production, making it a key target for immunological research.
When selecting a RAC2 antibody, consider the following methodological approach:
Application compatibility: Determine whether the antibody has been validated for your specific application (WB, IHC, IF, ELISA, FCM, IP, etc.)
Species reactivity: Ensure the antibody recognizes RAC2 in your species of interest (human, mouse, rat, etc.)
Epitope selection: For phosphorylation or activation state-specific studies, choose antibodies that recognize specific epitopes or conformations
Validation data: Review validation data including positive controls in tissues known to express RAC2 (lymphoid tissues, neutrophils)
Clone type: For detection of specific RAC2 variants or mutants, monoclonal antibodies may provide better specificity
Application | Purpose | Common Issues | Recommendation |
---|---|---|---|
Western Blot | Protein expression level | Cross-reactivity with RAC1/3 | Use antibodies targeting unique C-terminal region |
Immunohistochemistry | Tissue localization | Background in hematopoietic tissues | Optimize blocking and antibody dilution |
Immunofluorescence | Subcellular localization | Distinguishing active vs. inactive | Use activation-specific antibodies |
Flow Cytometry | Cell-specific expression | Fixation may affect epitope | Test multiple fixation protocols |
Immunoprecipitation | Protein-protein interactions | Low efficiency pull-down | Use high-affinity antibodies or tagged proteins |
Despite sharing approximately 92% amino acid sequence homology, RAC1 and RAC2 have distinct functions in cells. To distinguish between them:
Antibody selection: Use antibodies that target the C-terminal region where RAC1 and RAC2 differ significantly
Expression pattern analysis: RAC1 is ubiquitously expressed while RAC2 is predominantly in hematopoietic cells
Knockout validation: Use RAC2-knockout cells/tissues as negative controls to confirm antibody specificity
Isoform-specific primers: For mRNA analysis, design primers targeting unique regions
Functional assays: Utilize the differential roles of RAC1 vs. RAC2 in specific cellular functions
In B cells specifically, RAC2 deficiency has more pronounced effects on B cell development and immunological synapse formation compared to RAC1 deficiency, providing a functional readout to distinguish their roles .
RAC2 plays a critical role in B cell receptor (BCR)-mediated immunological synapse formation, which directly impacts antibody production. The mechanism involves:
BCR-triggered activation: Upon antigen recognition, BCR signaling activates RAC2 through guanine nucleotide exchange factors (GEFs) like DOCK2
Cytoskeletal remodeling: Activated RAC2 induces actin polymerization required for B cell spreading over antigen-presenting surfaces
Microcluster dynamics: RAC2 mediates the sustained growth of BCR microclusters at the B cell-antigen interface, crucial for signal amplification
Plasma cell differentiation: The DOCK2-RAC2 signaling axis is required for efficient plasma cell differentiation and subsequent antibody production
Research has demonstrated that RAC2-deficient B cells show impaired spreading responses and defective sustained growth of BCR microclusters. These cellular defects lead to severe impairment in plasma cell differentiation both in vitro and in vivo . Specifically, when DOCK2-deficient B cells (with impaired RAC2 activation) were stimulated with anti-IgM F(ab')2 antibody in the presence of IL-4 and IL-5, they failed to differentiate efficiently into plasma cells, highlighting the critical role of RAC2 activation in this process .
Several RAC2 mutations have been identified in patients with inborn errors of immunity, with distinct clinical and functional consequences:
E62K mutation: This gain-of-function mutation in the G3/Switch II domain is associated with myeloid deficit, altered neutrophil function, and lymphopenia
D63V mutation: Another gain-of-function mutation linked to juvenile myelomonocytic leukemia
G12R mutation: Discovered in patients with bone marrow hypoplasia and severe combined immunodeficiency syndrome with autosomal dominant inheritance; two G12R patients developed sepsis indicating significant impact on infection susceptibility
The molecular consequences of these mutations involve altered GTPase activity, disrupted effector binding, and changes in subcellular localization. When analyzing patient samples for these mutations, consider:
Using mutation-specific antibodies if available
Employing functional assays to assess RAC2 activation state
Analyzing downstream signaling pathways
Evaluating immune cell development and function
RAC2 expression and function have emerging significance in cancer biology, with potential applications in diagnosis and prognosis:
To utilize RAC2 as a cancer biomarker:
Employ immunohistochemistry to assess protein expression in tumor vs. normal tissues
Combine with other markers for increased diagnostic precision
Consider RAC2 genetic alterations in molecular profiling panels
Evaluate RAC2 expression in the context of tumor immune microenvironment
Advanced research is exploring how RAC2's role in immune cell function may influence tumor immunosurveillance and response to immunotherapies. The connection between RAC2 expression in tumor-infiltrating immune cells and cancer prognosis represents an important area for further investigation .
Detecting RAC2 activation presents technical challenges due to its highly regulated activity cycle. Recommended methodologies include:
RAC-GTP pull-down assays:
Use GST-PAK-CRIB domain fusion proteins to selectively bind active RAC2-GTP
Maintain cells at 37°C until lysis to preserve activation state
Include positive controls (GTPγS-treated lysates) and negative controls (GDP-treated lysates)
Rapid processing is essential as GTPase activity continues in lysates
Activation-specific antibodies:
Some antibodies can distinguish the GTP-bound active conformation
Validate with known RAC2 activators (e.g., chemokines in neutrophils)
Fix cells rapidly to preserve activation state
FRET-based biosensors:
For live-cell imaging of RAC2 activation dynamics
Requires optimization for each cell type
Provides spatial and temporal resolution of activation
Indirect readouts:
Phosphorylation of downstream targets (PAK1/2, LIMK)
Actin polymerization assays
Superoxide production (in neutrophils)
When troubleshooting, consider that serum starvation before stimulation can reduce baseline activation, and RAC2 activity is highly sensitive to temperature changes and mechanical stimulation during cell handling .
For optimal immunohistochemical detection of RAC2 in tissues:
Fixation optimization:
10% neutral buffered formalin (24-48 hours) works well for most tissues
For bone marrow/lymphoid tissues, consider shorter fixation times
Antigen retrieval:
Heat-induced epitope retrieval in citrate buffer (pH 6.0) is generally effective
For some epitopes, EDTA buffer (pH 9.0) may provide better results
Blocking strategy:
Include both protein block (5% BSA or normal serum) and peroxidase block
For lymphoid tissues with high endogenous biotin, use avidin/biotin blocking
Primary antibody incubation:
Optimize dilution (typically 1:100 to 1:500) based on antibody source
Incubate at 4°C overnight for best signal-to-noise ratio
Detection system:
Polymer-based detection systems often provide cleaner results than ABC methods
Consider tyramide signal amplification for low-abundance detection
Controls:
Include RAC2-high tissues (lymphoid tissues) as positive controls
Use RAC2-knockout tissues or isotype antibodies as negative controls
Research has shown that RAC2 protein expression can be effectively visualized in cancer and corresponding normal tissues using optimized IHC protocols. An approach using pressure cooker antigen thermal repair in citrate buffer followed by overnight incubation with RAC2 antibody at 4°C has proven successful .
Understanding RAC2's interactome is crucial for deciphering its functions. Recommended approaches include:
Co-immunoprecipitation (Co-IP):
Use activation state-specific conditions (GTPγS or GDP loading)
Consider crosslinking for transient interactions
Validate with reciprocal Co-IPs when possible
Use gentle lysis buffers to preserve complexes
Proximity labeling techniques:
BioID or APEX2 fusions to RAC2 for in vivo proximity labeling
Allows detection of transient or weak interactions
Requires careful controls to distinguish specific interactions
FRET/BRET-based interaction assays:
For real-time analysis of interactions in living cells
Can reveal spatial and temporal dynamics
Requires fluorescent protein fusions that may affect function
Pull-down assays with recombinant proteins:
Use purified RAC2 (WT, constitutively active, or dominant negative)
Load with specific nucleotides (GTPγS vs. GDP)
Pull down from cell lysates followed by mass spectrometry
Yeast two-hybrid screening:
Consider using constitutively active RAC2 as bait
Verify interactions with mammalian cell-based assays
When investigating the DOCK2-RAC2 interaction specifically, studies have successfully used pull-down assays with RAC2 loaded with different nucleotides to demonstrate the GEF activity of DOCK2's DHR-2 domain .
RAC2 antibodies have emerging utility in the evaluation of primary immunodeficiency disorders:
Diagnostic applications:
Assess RAC2 protein expression in patient lymphocytes and neutrophils
Evaluate RAC2 activation in response to stimuli (may be abnormal even with normal expression)
Identify specific RAC2 variants using mutation-specific antibodies (if available)
Functional evaluation:
Combine with functional assays of neutrophil chemotaxis, phagocytosis, and oxidative burst
Assess B cell immunological synapse formation using imaging with RAC2 antibodies
Evaluate downstream signaling pathway activation
Treatment monitoring:
Monitor reconstitution of RAC2 expression/function following hematopoietic stem cell transplantation
Assess normalization of immune cell development
Research has demonstrated that patients with RAC2 mutations (such as E62K, D63V, and G12R) present with distinct immunological phenotypes including altered neutrophil function, myeloid deficits, and severe combined immunodeficiency . When evaluating patients for potential RAC2-related immunodeficiencies, combining protein expression analysis with functional studies provides the most comprehensive assessment.
Based on pancancer analysis, RAC2 shows promise as a target in cancer immunotherapy research:
Prognostic significance:
Tumor immunity mechanisms:
RAC2 functions in immune cells that infiltrate tumors
Potentially modulates anti-tumor immune responses
May affect response to existing immunotherapies
Therapeutic approaches:
Small molecule inhibitors of RAC2 activation (e.g., targeting the DOCK2-RAC2 axis)
Antibody-drug conjugates targeting RAC2-expressing cells
Gene-edited cell therapies with enhanced RAC2 function
Combination therapy potential:
RAC2 modulation may sensitize tumors to immune checkpoint inhibitors
Could enhance efficacy of adoptive cell therapies
When investigating RAC2 in cancer contexts, researchers should consider both its direct effects on cancer cells and its functions in tumor-infiltrating immune cells, as both may contribute to disease progression and treatment response .
RAC2 functions as a molecular switch, cycling between an active GTP-bound state and an inactive GDP-bound state. This cycle is tightly regulated by guanine nucleotide exchange factors (GEFs), GTPase-activating proteins (GAPs), and guanine nucleotide dissociation inhibitors (GDIs) . When in its active state, RAC2 interacts with various effector proteins to regulate actin cytoskeleton remodeling, cell adhesion, migration, and other cellular behaviors .
RAC2 is predominantly expressed in immune cells, such as neutrophils, macrophages, and T lymphocytes. It is essential for the proper functioning of these cells, including their ability to migrate to sites of infection, produce reactive oxygen species, and phagocytose pathogens . Mutations in the RAC2 gene can lead to immunodeficiency disorders, highlighting its importance in maintaining immune system integrity .
Research has shown that RAC2 is involved in various pathological conditions, including cancer and immune disorders. For instance, RAC2 overexpression has been linked to the progression of certain cancers, making it a potential target for cancer therapy . Additionally, RAC2 mutations can cause primary immunodeficiency, leading to increased susceptibility to infections .
Mouse anti-human RAC2 antibodies are commonly used in research to study the expression and function of RAC2 in human cells. These antibodies are generated by immunizing mice with human RAC2 protein, leading to the production of specific antibodies that can be harvested and purified. These antibodies are valuable tools for various applications, including Western blotting, immunoprecipitation, and flow cytometry, allowing researchers to investigate RAC2’s role in different cellular contexts.