DOCK2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to DOCK2 Antibody

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.

BCR Signaling and B Cell Functions

  • 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 .

Antifungal Immunity

  • 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 .

Role in Studying Immune Responses

The DOCK2 Antibody enables researchers to:

  1. Map Signaling Pathways: Identify DOCK2-Rac interactions in leukocyte chemotaxis and phagocytosis.

  2. Assess Protein Expression: Quantify DOCK2 levels in immune cell subsets (e.g., B cells, macrophages) under inflammatory conditions.

  3. Validate Genetic Models: Confirm DOCK2 ablation in knockout mice or CRISPR-edited cell lines .

Implications in Disease Research

  • 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 .

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide.
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery time information.
Synonyms
DOCK2 antibody; KIAA0209Dedicator of cytokinesis protein 2 antibody
Target Names
Uniprot No.

Target Background

Function
DOCK2 plays a crucial role in cytoskeletal rearrangements essential for lymphocyte migration in response to chemokines. It activates RAC1 and RAC2, but not CDC42, by acting as a guanine nucleotide exchange factor (GEF). This function enables the exchange of bound GDP for free GTP. Additionally, DOCK2 may participate in IL2 transcriptional activation through the activation of RAC2.
Gene References Into Functions
  • A study demonstrated the applicability of the CPP-conjugation approach for developing novel anti-inflammatory drugs based on DOCK2 inhibition by investigating both cellular uptake and bioactivity. PMID: 28284862
  • DOCK2 has emerged as a potential therapeutic target for novel AML treatments due to its regulation of leukemia cell survival with elevated FLT3 activity. Moreover, DOCK2 sensitizes FLT3/ITD leukemic cells to conventional antileukemic agents. PMID: 27748370
  • Comparative genomics analysis revealed that SNPs within or proximal to CIITA (rs6498115), SMC6 (rs1834619), and KLHL29 (rs2288697) exhibited significant differentiation in the Amerindian-specific branch. Similarly, SNPs in ADAMTS9 (rs7631391), DOCK2 (rs77594147), SLC28A1 (rs28649017), ARHGAP5 (rs7151991), and CIITA (rs45601437) were prominent in the Asian comparison. PMID: 28100790
  • Autosomal recessive DOCK2 deficiency represents a novel Mendelian disorder characterized by pleiotropic defects affecting both hematopoietic and nonhematopoietic immunity. PMID: 26083206
  • Research findings have uncovered a previously unknown, nonredundant role for Elmo1 in regulating Dock2 levels and Dock2-dependent T cell migration in primary lymphocytes. PMID: 24821968
  • DOCK2 is essential for the normal migration and signal transduction of T and B cells. (Review) PMID: 23911989
  • DOCK2 mutations have been associated with esophageal adenocarcinoma. PMID: 23525077
  • Structural analysis revealed that the C-terminal Pro-rich tail of ELMO1 winds around the Src-homology 3 domain of DOCK2 to form an intermolecular 5-helix bundle. This intricate assembly of DOCK2 and ELMO1 regions creates a rigid structure that is crucial for their binding. PMID: 22331897
  • Studies have shown that CXCL13-mediated PCa cell invasion necessitates Akt and ERK12 activation, suggesting a novel role for DOCK2 in the proliferation of hormone-refractory CXCR5-positive PCa cells. PMID: 21645150
  • Prostate cancer cell lines exhibit differential expression of phosphoinositide-3 kinase (PI3K) catalytic subunit isoforms and dedicator of cytokinesis 2. PMID: 20412587
  • This research represents the first report to elucidate the prominent role of DOCK2 in hematopoietic malignancy. PMID: 20350533
  • DOCK2 mediates T cell receptor-induced activation of Rac2 and IL-2 transcription in Jurkat cells. PMID: 12176041
  • The association of DOCK2 with ELMO1 is critical for DOCK2-mediated Rac activation, suggesting that their association may serve as a therapeutic target for immunologic disorders caused by lymphocyte infiltration. PMID: 12829596
  • Nef binds the DOCK2-ELMO1 complex to activate rac and inhibit lymphocyte chemotaxis. PMID: 14737186
  • DOCK2 is required for efficient chemokine-stimulated lymphocyte attachment to VCAM-1 under shear stress. PMID: 17015707
  • DOCK2 and DOCK9 specifically recognize Rac2 and Cdc42 through their switch 1 and beta2-beta3 regions. The recognition mode via switch 1 appears to be conserved among diverse Rac-specific DHR-2 GEFs. PMID: 18056264
  • PI3K and Src-ELMO-Dock2 pathways operate in parallel to activate Rac2 and modulate chemotaxis in response to a CXCL8 gradient in neutrophils. PMID: 18662984
  • DOCK2 regulates microglial innate immunity independent of COX2 induction. Notably, DOCK2+ microglia are associated with human Alzheimer's disease pathology. PMID: 19729484

Show More

Hide All

Database Links

HGNC: 2988

OMIM: 603122

KEGG: hsa:1794

STRING: 9606.ENSP00000256935

UniGene: Hs.586174

Involvement In Disease
Immunodeficiency 40 (IMD40)
Protein Families
DOCK family
Subcellular Location
Endomembrane system; Peripheral membrane protein. Cytoplasm, cytoskeleton. Note=Colocalizes with F-actin.
Tissue Specificity
Specifically expressed in hematopoietic cells. Highly expressed in peripheral blood leukocytes, and expressed at intermediate level in thymus and spleen. Expressed at very low level in the small intestine and colon.

Q&A

What is DOCK2 and why is it important in immunological research?

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 .

What are the optimal cell types to use as positive controls for DOCK2 antibody validation?

For robust validation of DOCK2 antibodies, researchers should use:

Cell TypeCharacteristicsApplicationsReference
Jurkat cellsHuman acute T cell leukemia cell line with high DOCK2 expressionWB, IF/ICC
Ramos cellsHuman Burkitt's lymphoma B-cell lineWB
HL-60 cellsHuman promyelocytic leukemia cellsWB
Raji cellsHuman Burkitt's lymphoma cell lineWB
Primary lymphocytesIsolated from peripheral blood or lymphoid tissuesMultiple applications

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 .

What are the principal applications of DOCK2 antibodies in immunological research?

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.

How should researchers choose between monoclonal and polyclonal DOCK2 antibodies?

The selection between monoclonal and polyclonal DOCK2 antibodies should be based on specific research requirements:

Antibody TypeAdvantagesLimitationsBest ApplicationsExamples
Monoclonal- Consistent lot-to-lot reproducibility
- High specificity for single epitope
- Lower background in some applications
- May be sensitive to epitope modifications
- Potentially lower signal strength
- IP experiments
- Studies requiring high specificity
- Long-term projects needing consistency
MyBioSource DOCK2 Rabbit mAb
Santa Cruz DOCK2 Antibody (E-7)
Polyclonal- Recognize multiple epitopes
- More tolerant to protein denaturation
- Often provide stronger signal
- Batch-to-batch variation
- Potentially higher background
- Western blotting
- IHC of fixed tissues
- Detection of modified proteins
Abcam DOCK2 antibody
Proteintech DOCK2-Specific antibody

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 .

What validation steps are essential when using a new DOCK2 antibody?

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:

    • Use cell lines with known DOCK2 expression (Jurkat, Ramos, HL-60)

    • Include DOCK2-knockdown/knockout samples or non-hematopoietic cells as 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:

    • For WB: Optimize protein loading (50-100 μg recommended) and antibody dilution (1:500-1:2000)

    • For IHC: Test multiple fixation and antigen retrieval methods (heat-mediated with citrate buffer pH 6 recommended)

    • For IF/ICC: Optimize fixation, permeabilization, and antibody concentration (1:50-1:500)

  • Cross-reactivity assessment:

    • Test on samples from different species if cross-reactivity is claimed

    • Evaluate potential cross-reactivity with other DOCK family proteins

What are the methodological considerations for optimizing DOCK2 detection by Western blot?

Detection of DOCK2 by Western blot presents specific challenges due to its high molecular weight:

ChallengeSolutionTechnical Details
Poor transfer efficiency- Use lower percentage gels (6-8%)
- Optimize transfer conditions
- Extended transfer time (overnight at low voltage)
- Add 0.1% SDS to transfer buffer
- PVDF membrane recommended
Weak signal- Increase protein loading
- Optimize antibody concentration
- Load 50-100 μg total protein
- Try 1:500-1:2000 dilution range
- Extended primary antibody incubation (overnight at 4°C)
Multiple bands/degradation- Improve sample preparation
- Add fresh protease inhibitors
- Keep samples cold during preparation
- Use RIPA buffer with complete protease inhibitor cocktail
- Avoid repeated freeze-thaw cycles
High background- Optimize blocking and washing- Try 5% BSA instead of milk (especially for phospho-proteins)
- Increase wash duration (4-5 washes of 10 minutes)
- Use Tween-20 (0.1%) in wash buffer

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 .

How can DOCK2 antibodies be used to investigate B cell receptor signaling and plasma cell differentiation?

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:

    • Track DOCK2 expression during B cell to plasma cell transition using flow cytometry or Western blot

    • Combine with markers of plasma cell differentiation (CD138, Blimp-1)

    • Use DOCK2 inhibitors (e.g., CPYPP at 12.5 μM) to assess functional role

  • 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 .

What approaches enable the study of DOCK2's role in autoimmune and inflammatory diseases?

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:

    • In Sjögren's disease: Elevated DOCK2 expression in CD8+ T cells with amelioration of disease signs upon DOCK2 inhibition

    • In severe COVID-19: Decreased DOCK2 expression associated with disease severity and specific genetic variants

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.

How can researchers investigate DOCK2-Rac interactions in immune cell migration?

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 .

What are common causes for inconsistent results in DOCK2 immunofluorescence experiments?

Immunofluorescence detection of DOCK2 can present several technical challenges:

IssuePossible CausesMethodological Solutions
High background- Insufficient blocking
- Excessive antibody concentration
- Inadequate washing
- Extend blocking (1-2 hours or overnight)
- Titrate antibody (start 1:50-1:500)
- Increase wash steps (4-5 washes of 10 minutes)
Weak signal- Epitope masking
- Inadequate permeabilization
- Low DOCK2 expression
- Optimize antigen retrieval
- Adjust permeabilization (0.1-0.5% Triton X-100)
- Increase antibody concentration or incubation time
Variable staining patterns- Fixation artifacts
- Cell activation state differences
- Heterogeneous cell populations
- Standardize fixation protocol (4% PFA recommended)
- Control activation conditions
- Use cell-type specific markers for proper identification
Non-specific staining- Cross-reactivity
- Fc receptor binding
- Autofluorescence
- Include isotype controls
- Add Fc receptor blocking step
- Include DOCK2-deficient cells as negative control

For optimal results, use positive control cells like Jurkat with known DOCK2 expression , and include appropriate counterstains to provide context for DOCK2 localization.

How can cell-type specific differences in DOCK2 detection be addressed methodologically?

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:

    • Account for activation-dependent changes in DOCK2 expression

    • Consider cell-specific post-translational modifications that may affect epitope recognition

    • Be aware of potential isoform differences between cell types

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 .

What methodological approaches can improve DOCK2 co-immunoprecipitation experiments?

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:

    • Choose antibodies validated for IP applications (e.g., Bethyl Laboratories' Rabbit anti-DOCK2 Antibody)

    • Pre-clear lysates to reduce non-specific binding

    • Optimize antibody:lysate ratio and incubation conditions

  • 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 .

How can DOCK2 antibodies support research into novel therapeutic approaches?

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.

What methodology should be employed to investigate DOCK2's role in newly identified disease associations?

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.

How can single-cell techniques be integrated with DOCK2 antibodies for advanced immunological research?

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 Antibody: Research FAQs for Scientific Investigations

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.

What is DOCK2 and why is it important in immunological research?

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 .

What are the optimal cell types to use as positive controls for DOCK2 antibody validation?

For robust validation of DOCK2 antibodies, researchers should use:

Cell TypeCharacteristicsApplicationsReference
Jurkat cellsHuman acute T cell leukemia cell line with high DOCK2 expressionWB, IF/ICC
Ramos cellsHuman Burkitt's lymphoma B-cell lineWB
HL-60 cellsHuman promyelocytic leukemia cellsWB
Raji cellsHuman Burkitt's lymphoma cell lineWB
Primary lymphocytesIsolated from peripheral blood or lymphoid tissuesMultiple applications

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 .

What are the principal applications of DOCK2 antibodies in immunological research?

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.

How should researchers choose between monoclonal and polyclonal DOCK2 antibodies?

The selection between monoclonal and polyclonal DOCK2 antibodies should be based on specific research requirements:

Antibody TypeAdvantagesLimitationsBest ApplicationsExamples
Monoclonal- Consistent lot-to-lot reproducibility
- High specificity for single epitope
- Lower background in some applications
- May be sensitive to epitope modifications
- Potentially lower signal strength
- IP experiments
- Studies requiring high specificity
- Long-term projects needing consistency
MyBioSource DOCK2 Rabbit mAb
Santa Cruz DOCK2 Antibody (E-7)
Polyclonal- Recognize multiple epitopes
- More tolerant to protein denaturation
- Often provide stronger signal
- Batch-to-batch variation
- Potentially higher background
- Western blotting
- IHC of fixed tissues
- Detection of modified proteins
Abcam DOCK2 antibody
Proteintech DOCK2-Specific antibody

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 .

What validation steps are essential when using a new DOCK2 antibody?

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:

    • Use cell lines with known DOCK2 expression (Jurkat, Ramos, HL-60)

    • Include DOCK2-knockdown/knockout samples or non-hematopoietic cells as 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:

    • For WB: Optimize protein loading (50-100 μg recommended) and antibody dilution (1:500-1:2000)

    • For IHC: Test multiple fixation and antigen retrieval methods (heat-mediated with citrate buffer pH 6 recommended)

    • For IF/ICC: Optimize fixation, permeabilization, and antibody concentration (1:50-1:500)

  • Cross-reactivity assessment:

    • Test on samples from different species if cross-reactivity is claimed

    • Evaluate potential cross-reactivity with other DOCK family proteins

What are the methodological considerations for optimizing DOCK2 detection by Western blot?

Detection of DOCK2 by Western blot presents specific challenges due to its high molecular weight:

ChallengeSolutionTechnical Details
Poor transfer efficiency- Use lower percentage gels (6-8%)
- Optimize transfer conditions
- Extended transfer time (overnight at low voltage)
- Add 0.1% SDS to transfer buffer
- PVDF membrane recommended
Weak signal- Increase protein loading
- Optimize antibody concentration
- Load 50-100 μg total protein
- Try 1:500-1:2000 dilution range
- Extended primary antibody incubation (overnight at 4°C)
Multiple bands/degradation- Improve sample preparation
- Add fresh protease inhibitors
- Keep samples cold during preparation
- Use RIPA buffer with complete protease inhibitor cocktail
- Avoid repeated freeze-thaw cycles
High background- Optimize blocking and washing- Try 5% BSA instead of milk (especially for phospho-proteins)
- Increase wash duration (4-5 washes of 10 minutes)
- Use Tween-20 (0.1%) in wash buffer

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 .

How can DOCK2 antibodies be used to investigate B cell receptor signaling and plasma cell differentiation?

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:

    • Track DOCK2 expression during B cell to plasma cell transition using flow cytometry or Western blot

    • Combine with markers of plasma cell differentiation (CD138, Blimp-1)

    • Use DOCK2 inhibitors (e.g., CPYPP at 12.5 μM) to assess functional role

  • 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 .

What approaches enable the study of DOCK2's role in autoimmune and inflammatory diseases?

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:

    • In Sjögren's disease: Elevated DOCK2 expression in CD8+ T cells with amelioration of disease signs upon DOCK2 inhibition

    • In severe COVID-19: Decreased DOCK2 expression associated with disease severity and specific genetic variants

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.

How can researchers investigate DOCK2-Rac interactions in immune cell migration?

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 .

What are common causes for inconsistent results in DOCK2 immunofluorescence experiments?

Immunofluorescence detection of DOCK2 can present several technical challenges:

IssuePossible CausesMethodological Solutions
High background- Insufficient blocking
- Excessive antibody concentration
- Inadequate washing
- Extend blocking (1-2 hours or overnight)
- Titrate antibody (start 1:50-1:500)
- Increase wash steps (4-5 washes of 10 minutes)
Weak signal- Epitope masking
- Inadequate permeabilization
- Low DOCK2 expression
- Optimize antigen retrieval
- Adjust permeabilization (0.1-0.5% Triton X-100)
- Increase antibody concentration or incubation time
Variable staining patterns- Fixation artifacts
- Cell activation state differences
- Heterogeneous cell populations
- Standardize fixation protocol (4% PFA recommended)
- Control activation conditions
- Use cell-type specific markers for proper identification
Non-specific staining- Cross-reactivity
- Fc receptor binding
- Autofluorescence
- Include isotype controls
- Add Fc receptor blocking step
- Include DOCK2-deficient cells as negative control

For optimal results, use positive control cells like Jurkat with known DOCK2 expression , and include appropriate counterstains to provide context for DOCK2 localization.

How can cell-type specific differences in DOCK2 detection be addressed methodologically?

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:

    • Account for activation-dependent changes in DOCK2 expression

    • Consider cell-specific post-translational modifications that may affect epitope recognition

    • Be aware of potential isoform differences between cell types

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 .

What methodological approaches can improve DOCK2 co-immunoprecipitation experiments?

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:

    • Choose antibodies validated for IP applications (e.g., Bethyl Laboratories' Rabbit anti-DOCK2 Antibody)

    • Pre-clear lysates to reduce non-specific binding

    • Optimize antibody:lysate ratio and incubation conditions

  • 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 .

How can DOCK2 antibodies support research into novel therapeutic approaches?

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.

What methodology should be employed to investigate DOCK2's role in newly identified disease associations?

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.

How can single-cell techniques be integrated with DOCK2 antibodies for advanced immunological research?

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 .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.