The DOCK2 Antibody is a rabbit-derived polyclonal antibody designed to identify endogenous DOCK2 protein in human samples. It targets the ~200 kDa DOCK2 protein, a hematopoietic cell-specific guanine nucleotide exchange factor (GEF) that activates Rac GTPases to regulate immune cell migration, activation, and effector functions . This antibody is widely used in Western blotting (WB) and immunoprecipitation (IP) to investigate DOCK2's role in immune responses and disease mechanisms.
Mechanistic Insight: The antibody has been instrumental in identifying DOCK2 as the primary Rac activator downstream of B cell receptor (BCR) signaling. Studies in Dock2 knockout mice revealed impaired Rac1/Rac2 activation, leading to defective immunological synapse formation and reduced plasma cell differentiation .
Functional Validation: WB analyses using this antibody confirmed that DOCK2-deficient B cells exhibit attenuated proliferation and antibody production upon anti-IgM or CD40 stimulation .
Critical Role in Macrophages: Research employing the DOCK2 Antibody demonstrated that DOCK2-deficient macrophages fail to activate Rac GTPases, resulting in reduced reactive oxygen species (ROS) production and impaired fungal clearance (Candida albicans) .
Therapeutic Implications: Knockdown experiments in human PBMCs showed diminished proinflammatory cytokine responses (e.g., TNF-α, IL-6) to fungal components, highlighting DOCK2's conserved role in antifungal immunity .
The DOCK2 Antibody enables researchers to:
Map Signaling Pathways: Identify DOCK2-Rac interactions in leukocyte chemotaxis and phagocytosis.
Assess Protein Expression: Quantify DOCK2 levels in immune cell subsets (e.g., B cells, macrophages) under inflammatory conditions.
Validate Genetic Models: Confirm DOCK2 ablation in knockout mice or CRISPR-edited cell lines .
Autoimmunity: DOCK2 dysregulation is linked to aberrant B cell activation in autoimmune disorders. Antibody-based studies revealed reduced germinal center formation in Dock2 ⁻/⁻ mice, suggesting therapeutic targeting potential .
Infectious Diseases: The antibody has clarified DOCK2's role in antifungal defenses, with deficiencies correlating with increased susceptibility to candidiasis .
DOCK2 is a 211.9 kDa protein predominantly expressed in hematopoietic cells that functions as a guanine nucleotide exchange factor (GEF) for RAC1 and RAC2 small GTPases . It plays crucial roles in cytoskeletal rearrangements required for lymphocyte migration in response to chemokines, B cell receptor (BCR) signaling, and plasma cell differentiation . DOCK2 deficiency can lead to severe combined immunodeficiency characterized by early-onset invasive infections, highlighting its critical role in immune function . Recent research has implicated DOCK2 in various disease processes including autoimmune disorders like Sjögren's disease and severe COVID-19, making it an important target for immunological investigations .
For robust validation of DOCK2 antibodies, researchers should use:
When using these controls, researchers should verify DOCK2 detection at the expected molecular weight (approximately 200-212 kDa) and include appropriate negative controls such as non-hematopoietic cell lines or DOCK2-knockdown samples .
DOCK2 antibodies support multiple research applications:
Western Blotting (WB): Detecting DOCK2 protein expression levels in cell lysates, typically observed at 200-212 kDa
Immunohistochemistry (IHC): Visualizing DOCK2 distribution in tissue sections, particularly in lymphoid tissues
Immunofluorescence/Immunocytochemistry (IF/ICC): Examining subcellular localization of DOCK2, especially during immune cell activation and migration
Immunoprecipitation (IP): Isolating DOCK2 and its binding partners to study protein-protein interactions
ELISA: Quantitative detection of DOCK2 in experimental samples
Each application requires specific optimization of antibody concentration, sample preparation, and detection methods to achieve reliable results.
The selection between monoclonal and polyclonal DOCK2 antibodies should be based on specific research requirements:
For critical experiments, researchers should validate findings using both antibody types and consider the specific epitope location in relation to functional domains (DHR-1, DHR-2) of DOCK2 .
Comprehensive validation is crucial for reliable DOCK2 antibody-based experiments:
Molecular weight verification: Confirm detection at expected ~212 kDa by Western blot, noting that observed weights between 200-212 kDa have been reported
Positive and negative controls:
Specificity tests:
Blocking peptide competition assays
siRNA/shRNA knockdown to demonstrate signal reduction
Comparison with multiple antibodies targeting different DOCK2 epitopes
Application-specific optimization:
Cross-reactivity assessment:
Test on samples from different species if cross-reactivity is claimed
Evaluate potential cross-reactivity with other DOCK family proteins
Detection of DOCK2 by Western blot presents specific challenges due to its high molecular weight:
Example protocol: For detection of human DOCK2, PVDF membrane probed with 1 μg/mL of anti-DOCK2 antibody followed by HRP-conjugated secondary antibody under reducing conditions has shown consistent results .
DOCK2 plays a critical role in B cell function through regulation of Rac activation downstream of the B cell receptor (BCR). Methodological approaches include:
B cell activation studies:
Western blotting to monitor DOCK2 expression before and after BCR stimulation with anti-IgM F(ab')2 antibody
Immunofluorescence to track DOCK2 localization during BCR clustering and immunological synapse formation
Co-immunoprecipitation to identify DOCK2 interaction partners in the BCR signaling cascade
Plasma cell differentiation analysis:
Mechanistic investigations:
Rac activation assays to measure DOCK2-dependent Rac activity following BCR stimulation
Immunofluorescence to visualize B cell spreading and BCR microcluster growth
Antibody production assays comparing wild-type and DOCK2-deficient B cells
Research has shown that BCR-mediated Rac activation is almost completely lost in DOCK2-deficient B cells, resulting in impaired B cell spreading and sustained growth of BCR microclusters. Additionally, both in vitro and in vivo studies demonstrate that DOCK2 is required for efficient plasma cell differentiation and antigen-specific IgG antibody responses .
DOCK2 has been implicated in various autoimmune and inflammatory conditions. Key methodological strategies include:
Expression analysis in disease tissue:
Immunohistochemistry of affected tissues (e.g., salivary glands in Sjögren's disease)
Quantitative Western blot comparing DOCK2 levels in patient vs. healthy samples
Flow cytometry to assess DOCK2 expression in specific immune cell populations
Single-cell analysis approaches:
Combine DOCK2 antibodies with lineage markers for multi-parameter flow cytometry
Single-cell RNA-seq integration to correlate protein and transcript levels
Mass cytometry for high-dimensional analysis of DOCK2 in relation to other proteins
Functional studies with DOCK2 inhibition:
DOCK2 inhibitor (CPYPP) treatment in disease models
Monitor clinical parameters and immune cell function
Histological assessment of tissue inflammation
Disease-specific findings:
These approaches can help determine whether DOCK2 dysregulation is a cause or consequence of disease and evaluate its potential as a therapeutic target in autoimmune and inflammatory conditions.
Studying DOCK2's role as a guanine nucleotide exchange factor (GEF) for Rac is critical for understanding immune cell migration:
Biochemical interaction studies:
Co-immunoprecipitation with DOCK2 antibodies followed by Rac detection
Active Rac pull-down assays comparing wild-type and DOCK2-deficient cells
Domain-specific antibodies to understand the contribution of DHR-1 and DHR-2 domains
Live-cell imaging approaches:
Immunofluorescence to track DOCK2 translocation to the leading edge during migration
Co-localization studies with F-actin and phosphatidylinositol 3,4,5-triphosphate (PIP3)
Proximity ligation assay (PLA) to visualize endogenous DOCK2-Rac interactions
Functional migration assays:
Transwell migration assays comparing chemotaxis in presence/absence of DOCK2
3D matrix invasion assays with DOCK2 inhibition
Live-cell tracking of cell movement in response to chemoattractant gradients
Research has demonstrated that in neutrophils, DOCK2 regulates chemotaxis through PIP3-dependent membrane translocation and subsequent Rac activation. DOCK2-deficient neutrophils show abnormal migratory behavior with reduced translocation speed, impaired polarized accumulation of F-actin, and defective PIP3 localization at the leading edge .
Immunofluorescence detection of DOCK2 can present several technical challenges:
For optimal results, use positive control cells like Jurkat with known DOCK2 expression , and include appropriate counterstains to provide context for DOCK2 localization.
DOCK2 detection may vary across immune cell populations due to biological and technical factors:
Standardization approaches:
Use ratiometric measurements against housekeeping proteins
Implement consistent sample preparation protocols across cell types
Validate with multiple detection methods (flow cytometry, Western blot, immunofluorescence)
Cell-type optimization:
Adjust permeabilization conditions based on cell size and membrane composition
Optimize antibody concentration for each cell type
Consider cell-specific fixation requirements
Controls and normalization:
Include relevant positive controls for each cell population
Use cell-type specific markers to normalize DOCK2 detection
Employ multiple antibodies targeting different DOCK2 epitopes
Biological considerations:
Research has shown cell-type specific regulation of DOCK2, including differential expression between lymphocytes and myeloid cells, and cell-specific effects of genetic variants on DOCK2 expression .
Co-immunoprecipitation (co-IP) of DOCK2 requires careful optimization due to its large size and role in protein complexes:
Lysis condition optimization:
Use gentle lysis buffers (NP-40 or CHAPS-based) to preserve interactions
Include protease and phosphatase inhibitors
Maintain cold temperature throughout sample preparation
Antibody selection and protocol:
Control experiments:
Include isotype control antibodies
Use DOCK2-deficient cells as negative controls
Include known DOCK2 interactors as positive controls
Detection optimization:
Use gradient gels for better separation of high molecular weight proteins
Consider silver staining followed by mass spectrometry for unbiased identification
Optimize Western blot detection of co-precipitated proteins
Interaction validation:
Perform reverse co-IP where possible
Use domain mutants to map interaction regions
Consider proximity ligation assay as complementary approach
When studying DOCK2-Rac interactions, researchers should consider using antibodies against activated Rac and include proper controls to distinguish specific from non-specific interactions .
DOCK2 antibodies can facilitate research into therapeutic targeting of DOCK2-dependent pathways:
Target validation studies:
Use antibodies to monitor DOCK2 expression in disease models before and after treatment
Correlate DOCK2 levels with disease progression and severity
Compare with effects of small molecule DOCK2 inhibitors like CPYPP
Biomarker development:
Quantify DOCK2 expression levels in patient samples as potential biomarkers
Correlate with disease activity and treatment response
Develop standardized immunoassays for clinical research
Mechanism-of-action studies:
Track DOCK2 localization and interaction changes during therapeutic intervention
Measure downstream effects on Rac activation and cytoskeletal reorganization
Monitor changes in immune cell migration and function
Emerging therapeutic areas:
Sjögren's disease: DOCK2 inhibition with CPYPP ameliorated disease signs in mouse models
Severe COVID-19: DOCK2 expression was suppressed in severe cases, suggesting potential therapeutic approaches targeting DOCK2 pathways
Hematopoietic malignancies: DOCK2's selective expression in hematopoietic cells makes it a potential therapeutic target
DOCK2 antibodies can serve as critical tools for validating this pathway as a therapeutic target and developing companion diagnostics for future targeted therapies.
As DOCK2 is implicated in an expanding range of diseases, systematic research approaches include:
Expression profiling workflow:
Initial screening with immunohistochemistry of affected tissues
Quantification by Western blot comparing disease vs. healthy samples
Single-cell analysis to identify cell-specific abnormalities
Functional characterization:
Assess impact of disease-associated variants on DOCK2 expression and function
Compare Rac activation in patient-derived cells vs. controls
Evaluate downstream effects on cell migration and immune function
Disease model development:
Generate relevant cellular or animal models incorporating disease-specific DOCK2 alterations
Validate models using DOCK2 antibodies to confirm appropriate expression patterns
Test DOCK2 inhibitors (e.g., CPYPP) for therapeutic potential
Translational research approach:
Correlate DOCK2 expression with clinical parameters
Evaluate potential as biomarker through standardized immunoassays
Assess genetic variations affecting DOCK2 expression or function
Recent research has identified DOCK2's involvement in diverse conditions including severe COVID-19 and Sjögren's disease , highlighting the importance of systematic investigation of its role in newly identified disease associations.
Integration of DOCK2 antibodies with single-cell technologies enables sophisticated analysis of immune function:
Single-cell protein analysis:
Flow cytometry and mass cytometry (CyTOF) with intracellular DOCK2 staining
Imaging mass cytometry for spatial resolution of DOCK2 expression
Spectral flow cytometry for high-parameter analysis with reduced compensation issues
Multi-omic approaches:
CITE-seq combining antibody detection with transcriptomics
Spatial transcriptomics with protein detection for tissue context
Single-cell proteomics with DOCK2 antibodies for protein network analysis
Functional single-cell assays:
Live-cell imaging of individual cell migration with DOCK2 visualization
Correlation of DOCK2 expression with functional readouts at single-cell level
Microfluidic approaches to study DOCK2-dependent migration
Clinical applications:
Immune monitoring in clinical trials targeting DOCK2-dependent pathways
Patient stratification based on DOCK2 expression patterns
Correlation of treatment response with DOCK2 levels in specific cell populations
Research has demonstrated the value of these approaches, with single-cell RNA-sequencing identifying cell-type-specific downregulation of DOCK2 in COVID-19, particularly in non-classical monocytes , and revealing elevated DOCK2 expression in specific T cell populations in Sjögren's disease .
DOCK2 (Dedicator of cytokinesis 2) is a critical protein in immune cell function, serving as a guanine nucleotide exchange factor (GEF) that activates Rac1 and Rac2 small GTPases. This comprehensive FAQ document addresses common questions researchers encounter when working with DOCK2 antibodies in academic research settings.
DOCK2 is a 211.9 kDa protein predominantly expressed in hematopoietic cells that functions as a guanine nucleotide exchange factor (GEF) for RAC1 and RAC2 small GTPases . It plays crucial roles in cytoskeletal rearrangements required for lymphocyte migration in response to chemokines, B cell receptor (BCR) signaling, and plasma cell differentiation . DOCK2 deficiency can lead to severe combined immunodeficiency characterized by early-onset invasive infections, highlighting its critical role in immune function . Recent research has implicated DOCK2 in various disease processes including autoimmune disorders like Sjögren's disease and severe COVID-19, making it an important target for immunological investigations .
For robust validation of DOCK2 antibodies, researchers should use:
When using these controls, researchers should verify DOCK2 detection at the expected molecular weight (approximately 200-212 kDa) and include appropriate negative controls such as non-hematopoietic cell lines or DOCK2-knockdown samples .
DOCK2 antibodies support multiple research applications:
Western Blotting (WB): Detecting DOCK2 protein expression levels in cell lysates, typically observed at 200-212 kDa
Immunohistochemistry (IHC): Visualizing DOCK2 distribution in tissue sections, particularly in lymphoid tissues
Immunofluorescence/Immunocytochemistry (IF/ICC): Examining subcellular localization of DOCK2, especially during immune cell activation and migration
Immunoprecipitation (IP): Isolating DOCK2 and its binding partners to study protein-protein interactions
ELISA: Quantitative detection of DOCK2 in experimental samples
Each application requires specific optimization of antibody concentration, sample preparation, and detection methods to achieve reliable results.
The selection between monoclonal and polyclonal DOCK2 antibodies should be based on specific research requirements:
For critical experiments, researchers should validate findings using both antibody types and consider the specific epitope location in relation to functional domains (DHR-1, DHR-2) of DOCK2 .
Comprehensive validation is crucial for reliable DOCK2 antibody-based experiments:
Molecular weight verification: Confirm detection at expected ~212 kDa by Western blot, noting that observed weights between 200-212 kDa have been reported
Positive and negative controls:
Specificity tests:
Blocking peptide competition assays
siRNA/shRNA knockdown to demonstrate signal reduction
Comparison with multiple antibodies targeting different DOCK2 epitopes
Application-specific optimization:
Cross-reactivity assessment:
Test on samples from different species if cross-reactivity is claimed
Evaluate potential cross-reactivity with other DOCK family proteins
Detection of DOCK2 by Western blot presents specific challenges due to its high molecular weight:
Example protocol: For detection of human DOCK2, PVDF membrane probed with 1 μg/mL of anti-DOCK2 antibody followed by HRP-conjugated secondary antibody under reducing conditions has shown consistent results .
DOCK2 plays a critical role in B cell function through regulation of Rac activation downstream of the B cell receptor (BCR). Methodological approaches include:
B cell activation studies:
Western blotting to monitor DOCK2 expression before and after BCR stimulation with anti-IgM F(ab')2 antibody
Immunofluorescence to track DOCK2 localization during BCR clustering and immunological synapse formation
Co-immunoprecipitation to identify DOCK2 interaction partners in the BCR signaling cascade
Plasma cell differentiation analysis:
Mechanistic investigations:
Rac activation assays to measure DOCK2-dependent Rac activity following BCR stimulation
Immunofluorescence to visualize B cell spreading and BCR microcluster growth
Antibody production assays comparing wild-type and DOCK2-deficient B cells
Research has shown that BCR-mediated Rac activation is almost completely lost in DOCK2-deficient B cells, resulting in impaired B cell spreading and sustained growth of BCR microclusters. Additionally, both in vitro and in vivo studies demonstrate that DOCK2 is required for efficient plasma cell differentiation and antigen-specific IgG antibody responses .
DOCK2 has been implicated in various autoimmune and inflammatory conditions. Key methodological strategies include:
Expression analysis in disease tissue:
Immunohistochemistry of affected tissues (e.g., salivary glands in Sjögren's disease)
Quantitative Western blot comparing DOCK2 levels in patient vs. healthy samples
Flow cytometry to assess DOCK2 expression in specific immune cell populations
Single-cell analysis approaches:
Combine DOCK2 antibodies with lineage markers for multi-parameter flow cytometry
Single-cell RNA-seq integration to correlate protein and transcript levels
Mass cytometry for high-dimensional analysis of DOCK2 in relation to other proteins
Functional studies with DOCK2 inhibition:
DOCK2 inhibitor (CPYPP) treatment in disease models
Monitor clinical parameters and immune cell function
Histological assessment of tissue inflammation
Disease-specific findings:
These approaches can help determine whether DOCK2 dysregulation is a cause or consequence of disease and evaluate its potential as a therapeutic target in autoimmune and inflammatory conditions.
Studying DOCK2's role as a guanine nucleotide exchange factor (GEF) for Rac is critical for understanding immune cell migration:
Biochemical interaction studies:
Co-immunoprecipitation with DOCK2 antibodies followed by Rac detection
Active Rac pull-down assays comparing wild-type and DOCK2-deficient cells
Domain-specific antibodies to understand the contribution of DHR-1 and DHR-2 domains
Live-cell imaging approaches:
Immunofluorescence to track DOCK2 translocation to the leading edge during migration
Co-localization studies with F-actin and phosphatidylinositol 3,4,5-triphosphate (PIP3)
Proximity ligation assay (PLA) to visualize endogenous DOCK2-Rac interactions
Functional migration assays:
Transwell migration assays comparing chemotaxis in presence/absence of DOCK2
3D matrix invasion assays with DOCK2 inhibition
Live-cell tracking of cell movement in response to chemoattractant gradients
Research has demonstrated that in neutrophils, DOCK2 regulates chemotaxis through PIP3-dependent membrane translocation and subsequent Rac activation. DOCK2-deficient neutrophils show abnormal migratory behavior with reduced translocation speed, impaired polarized accumulation of F-actin, and defective PIP3 localization at the leading edge .
Immunofluorescence detection of DOCK2 can present several technical challenges:
For optimal results, use positive control cells like Jurkat with known DOCK2 expression , and include appropriate counterstains to provide context for DOCK2 localization.
DOCK2 detection may vary across immune cell populations due to biological and technical factors:
Standardization approaches:
Use ratiometric measurements against housekeeping proteins
Implement consistent sample preparation protocols across cell types
Validate with multiple detection methods (flow cytometry, Western blot, immunofluorescence)
Cell-type optimization:
Adjust permeabilization conditions based on cell size and membrane composition
Optimize antibody concentration for each cell type
Consider cell-specific fixation requirements
Controls and normalization:
Include relevant positive controls for each cell population
Use cell-type specific markers to normalize DOCK2 detection
Employ multiple antibodies targeting different DOCK2 epitopes
Biological considerations:
Research has shown cell-type specific regulation of DOCK2, including differential expression between lymphocytes and myeloid cells, and cell-specific effects of genetic variants on DOCK2 expression .
Co-immunoprecipitation (co-IP) of DOCK2 requires careful optimization due to its large size and role in protein complexes:
Lysis condition optimization:
Use gentle lysis buffers (NP-40 or CHAPS-based) to preserve interactions
Include protease and phosphatase inhibitors
Maintain cold temperature throughout sample preparation
Antibody selection and protocol:
Control experiments:
Include isotype control antibodies
Use DOCK2-deficient cells as negative controls
Include known DOCK2 interactors as positive controls
Detection optimization:
Use gradient gels for better separation of high molecular weight proteins
Consider silver staining followed by mass spectrometry for unbiased identification
Optimize Western blot detection of co-precipitated proteins
Interaction validation:
Perform reverse co-IP where possible
Use domain mutants to map interaction regions
Consider proximity ligation assay as complementary approach
When studying DOCK2-Rac interactions, researchers should consider using antibodies against activated Rac and include proper controls to distinguish specific from non-specific interactions .
DOCK2 antibodies can facilitate research into therapeutic targeting of DOCK2-dependent pathways:
Target validation studies:
Use antibodies to monitor DOCK2 expression in disease models before and after treatment
Correlate DOCK2 levels with disease progression and severity
Compare with effects of small molecule DOCK2 inhibitors like CPYPP
Biomarker development:
Quantify DOCK2 expression levels in patient samples as potential biomarkers
Correlate with disease activity and treatment response
Develop standardized immunoassays for clinical research
Mechanism-of-action studies:
Track DOCK2 localization and interaction changes during therapeutic intervention
Measure downstream effects on Rac activation and cytoskeletal reorganization
Monitor changes in immune cell migration and function
Emerging therapeutic areas:
Sjögren's disease: DOCK2 inhibition with CPYPP ameliorated disease signs in mouse models
Severe COVID-19: DOCK2 expression was suppressed in severe cases, suggesting potential therapeutic approaches targeting DOCK2 pathways
Hematopoietic malignancies: DOCK2's selective expression in hematopoietic cells makes it a potential therapeutic target
DOCK2 antibodies can serve as critical tools for validating this pathway as a therapeutic target and developing companion diagnostics for future targeted therapies.
As DOCK2 is implicated in an expanding range of diseases, systematic research approaches include:
Expression profiling workflow:
Initial screening with immunohistochemistry of affected tissues
Quantification by Western blot comparing disease vs. healthy samples
Single-cell analysis to identify cell-specific abnormalities
Functional characterization:
Assess impact of disease-associated variants on DOCK2 expression and function
Compare Rac activation in patient-derived cells vs. controls
Evaluate downstream effects on cell migration and immune function
Disease model development:
Generate relevant cellular or animal models incorporating disease-specific DOCK2 alterations
Validate models using DOCK2 antibodies to confirm appropriate expression patterns
Test DOCK2 inhibitors (e.g., CPYPP) for therapeutic potential
Translational research approach:
Correlate DOCK2 expression with clinical parameters
Evaluate potential as biomarker through standardized immunoassays
Assess genetic variations affecting DOCK2 expression or function
Recent research has identified DOCK2's involvement in diverse conditions including severe COVID-19 and Sjögren's disease , highlighting the importance of systematic investigation of its role in newly identified disease associations.
Integration of DOCK2 antibodies with single-cell technologies enables sophisticated analysis of immune function:
Single-cell protein analysis:
Flow cytometry and mass cytometry (CyTOF) with intracellular DOCK2 staining
Imaging mass cytometry for spatial resolution of DOCK2 expression
Spectral flow cytometry for high-parameter analysis with reduced compensation issues
Multi-omic approaches:
CITE-seq combining antibody detection with transcriptomics
Spatial transcriptomics with protein detection for tissue context
Single-cell proteomics with DOCK2 antibodies for protein network analysis
Functional single-cell assays:
Live-cell imaging of individual cell migration with DOCK2 visualization
Correlation of DOCK2 expression with functional readouts at single-cell level
Microfluidic approaches to study DOCK2-dependent migration
Clinical applications:
Immune monitoring in clinical trials targeting DOCK2-dependent pathways
Patient stratification based on DOCK2 expression patterns
Correlation of treatment response with DOCK2 levels in specific cell populations
Research has demonstrated the value of these approaches, with single-cell RNA-sequencing identifying cell-type-specific downregulation of DOCK2 in COVID-19, particularly in non-classical monocytes , and revealing elevated DOCK2 expression in specific T cell populations in Sjögren's disease .