SRGAP3 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Applications

SRGAP3 HRP-conjugated antibodies are polyclonal or monoclonal antibodies chemically linked to HRP, facilitating target protein detection via substrate conversion (e.g., TMB or DAB) .

Key ApplicationsDetails
ELISAQuantification of SRGAP3 in biological samples (e.g., CSB-PA022663LB01HU) .
Western Blot (WB)Detection of SRGAP3 at ~125 kDa in human, mouse, and rat brain tissues .
Immunohistochemistry (IHC)Localization in human ovary cancer tissues (validated with citrate/TE buffer antigen retrieval) .

Table 2: Validation Data (Proteintech 29624-1-AP)

ApplicationSampleDilutionResult
WBMouse brain tissue1:2000–1:12000Clear band at 125 kDa
IHCHuman ovary cancer1:500–1:2000Cytoplasmic and membrane staining

Research Findings

  • Neuronal Differentiation: SRGAP3 regulates Rac1-dependent neurite outgrowth in Neuro2A cells. Overexpression inhibits differentiation, while knockdown enhances it .

  • Subcellular Localization: In undifferentiated Neuro2A cells, endogenous SRGAP3 localizes to the cytoplasm and nucleus. During differentiation, it redistributes to neurites and the plasma membrane .

  • Antibody Specificity: Polyclonal antibodies (e.g., 3A1, 3A3) show no cross-reactivity with srGAP1 or srGAP2 in Western blots, confirming specificity for SRGAP3 .

Technical Considerations

  • Buffer Compatibility: Conjugation efficiency depends on antibody buffer composition; additives like azides or amines may interfere .

  • Storage: HRP conjugates require light-protected storage at 4°C (short-term) or -20°C with 50% glycerol (long-term) .

  • Controls: Use GFP-tagged SRGAP3-transfected lysates (positive) and non-transfected cells (negative) for WB validation .

Supplier Comparison

  • Cusabio: Focuses on ELISA applications with full-length SRGAP3 recognition .

  • Aviva SysBio: Targets the N-terminal region (AA 1-50) and validates for WB across multiple species .

  • Proteintech: Provides broad application validation (WB, IHC, IF) with epitope coverage across human, mouse, and rat samples .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
ARHGAP 14 antibody; ARHGAP14 antibody; Gbi antibody; ME GAP antibody; MEGAP antibody; Mental disorder associated GAP antibody; Mental disorder-associated GAP antibody; Rho GTPase activating protein 14 antibody; Rho GTPase-activating protein 14 antibody; SLIT ROBO Rho GTPase activating protein 3 antibody; SLIT-ROBO Rho GTPase-activating protein 3 antibody; srGAP 2 antibody; srGAP 3 antibody; srGAP2 antibody; srGAP3 antibody; SRGP 2 antibody; SRGP2 antibody; SRGP3_HUMAN antibody; WAVE associated Rac GTPase activating protein antibody; WAVE-associated Rac GTPase-activating protein antibody; WRP antibody
Target Names
SRGAP3
Uniprot No.

Target Background

Function

SRGAP3 is a GTPase-activating protein (GAP) for RAC1 and possibly Cdc42, but not for RhoA. It may modulate RAC1 signaling in neurons.

Gene References Into Functions

SRGAP3 Function and Interactions:

  1. A single PXXP motif in srGAP3's C-terminus mediates binding to multiple SH3 domains. (PMID: 25819436)
  2. Nuclear srGAP3 interacts with Brg1 via its C-terminus and Brg1's ATPase motif. (PMID: 24561795)
  3. SRGAP3 haploinsufficiency is strongly implicated in a patient's mental retardation phenotype. (PMID: 24300292)
  4. srGAP3 exhibits tumor suppressor-like activity in human mammary epithelial cells (HMECs), likely by negatively regulating Rac1. (PMID: 23108406)
  5. SRGAP3 is a putative factor in severe mental retardation. (PMID: 12195014)
  6. MEGAP (a related protein) negatively regulates cell migration by affecting the actin and microtubule cytoskeleton and focal complex formation. (PMID: 16730001)
  7. No association was found between SRGAP3/MEGAP haploinsufficiency and mental retardation in a specific study. (PMID: 19433673)
  8. SRGAP3 is considered a major determinant of mental retardation in distal 3p deletions. (PMID: 19760623)
  9. The FNBP2 family (FNBP2, ARHGAP13, ARHGAP14, ARHGAP4) is characterized by FCH, RhoGAP, and SH3 domains. (PMID: 12736724)

Database Links

HGNC: 19744

OMIM: 606525

KEGG: hsa:9901

STRING: 9606.ENSP00000373347

UniGene: Hs.654743

Involvement In Disease
A chromosomal aberration involving SRGAP3 is found in a patient with severe idiopathic mental retardation (PubMed:12195014). Translocation t(X;3)(p11.2;p25) (PubMed:12195014).
Tissue Specificity
Highly expressed in adult and fetal brain. Expressed at low levels in kidney. Isoform 3 is expressed in the kidney but is absent in the brain.

Q&A

What is SRGAP3 and why is it a significant target for antibody-based research?

SRGAP3 (SLIT-ROBO Rho GTPase Activating Protein 3) functions as a GTPase-activating protein primarily for RAC1 and potentially Cdc42, but not for RhoA small GTPase. It plays a critical role in attenuating RAC1 signaling in neurons, making it an important target for neurodevelopmental and neurological research . The protein is also known by several alternative names including ARHGAP14, MEGAP, KIAA0411, WRP (WAVE-associated Rac GTPase-activating protein), and Mental disorder-associated GAP, indicating its multifaceted roles in cellular processes and potential implications in pathological conditions . Using specific antibodies against SRGAP3 enables researchers to investigate its expression, localization, and function in various experimental contexts.

How does the molecular structure of SRGAP3 influence antibody selection for neurological research?

SRGAP3 is a large protein with a calculated molecular weight of 124 kDa and an observed molecular weight of approximately 125 kDa in SDS-PAGE . When selecting antibodies for neurological research, it's crucial to consider which functional domains you need to target. Different antibodies recognize distinct epitopes within the protein, such as AA 201-300, AA 709-955, AA 900-1000, or N-terminal regions . For studying SRGAP3's role in neuronal RAC1 signaling, antibodies targeting the GTPase-activating domain would be most appropriate. Selecting antibodies validated in brain tissue is also essential, as demonstrated by successful detection in mouse and rat brain tissues .

How should I design experiments to compare SRGAP3 expression in different brain regions using HRP-conjugated antibodies?

For comparative analysis of SRGAP3 expression across brain regions, implement a systematic approach that accounts for regional variability. Begin with tissue preparation using consistent fixation protocols—paraformaldehyde fixation followed by paraffin embedding works effectively for SRGAP3 detection . For immunohistochemistry, heat-mediated antigen retrieval with citrate buffer (pH 6.0) is recommended based on validated protocols . Use serial dilutions (1:500-1:2000) of the HRP-conjugated antibody to determine optimal concentration for each brain region . Include positive controls such as cerebral cortex tissue where SRGAP3 expression has been confirmed and negative controls like colon tissue which shows low expression . For quantitative analysis, implement digital image analysis with normalization to housekeeping proteins, and validate findings with alternative methods such as qRT-PCR or western blotting.

What control samples should be included when using SRGAP3 antibodies for immunohistochemistry studies?

A robust control strategy for SRGAP3 immunohistochemistry should include multiple levels of validation. Positive tissue controls should include cerebral cortex, fallopian tube, and urinary bladder tissues, which have demonstrated successful staining with SRGAP3 antibodies . Negative controls should include colon tissue, which shows expected low expression of SRGAP3 . Technical negative controls must be included by omitting the primary antibody while maintaining identical protocol conditions. For antibody validation, consider using tissues from SRGAP3 knockout models or SRGAP3-knockdown cells if available. When evaluating staining patterns, lymph node tissue can serve as an additional reference point . For HRP-conjugated antibodies specifically, include enzyme inhibition controls to account for potential endogenous peroxidase activity.

How do I determine the optimal dilution for SRGAP3 HRP-conjugated antibodies in various applications?

Determining optimal dilution requires systematic titration across applications. For ELISA applications, begin with a broad range titration (1:500, 1:1000, 1:2000, 1:5000) of the HRP-conjugated SRGAP3 antibody, then narrow to find the concentration that provides maximum specific signal with minimal background . For immunohistochemistry, start with the manufacturer's recommended dilution range (1:500-1:2000) and prepare a dilution series on identical tissue sections . For western blotting, the recommended range of 1:2000-1:12000 provides a starting point, but sample-dependent optimization is essential . Create a standardized signal-to-noise assessment protocol where signal intensity from known SRGAP3-positive regions is compared to background areas. Document optimization results in a table format recording antibody dilution, incubation time, temperature, and buffer conditions for reproducibility.

What protocol modifications are necessary when using HRP-conjugated SRGAP3 antibodies versus unconjugated primary antibodies?

When transitioning from unconjugated to HRP-conjugated SRGAP3 antibodies, several protocol modifications are essential. First, eliminate the secondary antibody incubation step entirely, as the HRP is directly coupled to the primary antibody . Reduce the primary antibody incubation time by approximately 25% compared to unconjugated antibodies, as direct detection systems often reach saturation more quickly. Implement more stringent blocking procedures using 3-5% BSA or commercial blocking buffers to minimize background—particularly important since you lose the specificity advantage of the two-antibody system . For wash steps, increase both duration and number (minimum 5 washes of 5 minutes each) to remove unbound conjugated antibody effectively. When developing the signal, reduce substrate incubation time by 30-50% compared to two-step detection to prevent oversaturation and maintain signal linearity. HRP-conjugated antibodies are also more sensitive to storage conditions, so aliquot and store at -20°C, avoiding repeated freeze-thaw cycles and exposure to light .

How should antigen retrieval protocols be modified for optimal detection of SRGAP3 in different tissue types?

Successful SRGAP3 detection requires tissue-specific antigen retrieval optimization. Heat-mediated antigen retrieval with citrate buffer (pH 6.0) has been validated for multiple tissue types including fallopian tube, urinary bladder, cerebral cortex, lymph node, and colon tissues . For neuronal tissues with high lipid content, consider extending the antigen retrieval duration by 5-10 minutes beyond standard protocols. In tissues with dense extracellular matrix (such as connective tissues), a dual approach combining protease treatment (0.05% trypsin for 10-15 minutes at 37°C) followed by heat-mediated retrieval may improve epitope accessibility. For formalin-fixed tissues with extended fixation times, stronger retrieval conditions using Tris-EDTA buffer (pH 9.0) may be necessary . Monitor retrieval efficiency by comparing signal intensity and background levels across different conditions. Document optimal conditions for each tissue type in a standardized format to ensure reproducibility across experiments.

What storage conditions maximize the shelf life and activity of HRP-conjugated SRGAP3 antibodies?

To preserve HRP-conjugated SRGAP3 antibody functionality, implement a comprehensive storage strategy. Store the antibody at -20°C in manufacturer-provided buffer containing 50% glycerol and stabilizers . Upon receipt, immediately prepare small working aliquots (5-20 μL) to minimize freeze-thaw cycles—each cycle can reduce activity by approximately 10-15% . Protect from light exposure, as HRP is photosensitive and can lose activity when repeatedly exposed to light. When removing from storage, thaw aliquots quickly at room temperature and keep on ice during experiment preparation. Add reducing agents (2-mercaptoethanol at 20 mM final concentration) to working dilutions just prior to use to protect the HRP active site. For long-term storage beyond six months, consider adding protease inhibitors to prevent degradation. Under optimal storage conditions and with proper handling, HRP-conjugated antibodies maintain >90% activity for at least 12 months .

How can SRGAP3 antibodies be utilized to investigate the interaction between SRGAP3 and RAC1 signaling in neuronal development?

Investigating SRGAP3-RAC1 interactions in neuronal development requires a multi-technique approach. Begin with co-immunoprecipitation studies using SRGAP3 antibodies to pull down protein complexes, followed by immunoblotting for RAC1 and its activated form . For spatial analysis, perform dual immunofluorescence labeling with the SRGAP3 antibody alongside RAC1 antibodies in developing neural tissues or neuronal cultures . Implement proximity ligation assays (PLA) to visualize and quantify direct SRGAP3-RAC1 interactions at the subcellular level. To establish functional relationships, combine SRGAP3 knockdown or overexpression with live cell imaging using FRET-based RAC1 activity biosensors. For in vivo studies, utilize SRGAP3 antibodies for immunohistochemistry in brain sections from developmental timepoints, correlating SRGAP3 expression patterns with known RAC1 activity zones . Supplement antibody-based approaches with biochemical GTPase activity assays to directly measure how SRGAP3 modulates RAC1 activation states in response to guidance cues like SLIT proteins.

What strategies should be employed to differentiate between SRGAP3 and other SRGAP family members in experimental systems?

Differentiation between SRGAP family members requires careful antibody selection and validation. First, perform sequence alignment analysis to identify unique regions in SRGAP3 compared to SRGAP1 and SRGAP2. Select antibodies targeting epitopes with minimal sequence homology, such as those recognizing the C-terminal domain (AA 956-989) where divergence is greatest . Validate antibody specificity through western blot analysis comparing recombinant SRGAP proteins and using cells with CRISPR knockout of specific SRGAP members. Implement peptide competition assays using synthetic peptides corresponding to the epitope regions of each SRGAP protein. For immunohistochemistry applications, compare staining patterns with published expression data for each family member, noting that SRGAP3 shows distinctive expression in cerebral cortex while having low expression in colon tissue . Complement antibody-based approaches with mRNA detection methods like RNAscope to independently confirm isoform-specific expression patterns.

How can SRGAP3 HRP-conjugated antibodies be adapted for high-throughput screening of potential neurological therapeutic targets?

Adapting SRGAP3 HRP-conjugated antibodies for high-throughput screening requires systematic protocol optimization. Develop a miniaturized ELISA format in 384-well plates using robotic liquid handling for consistent antibody application at standardized concentrations (typically 1:1000 dilution) . Implement a signal amplification system such as tyramide signal amplification to enhance detection sensitivity while maintaining specificity. Create a reference standard curve using recombinant SRGAP3 protein to enable quantitative analysis across plate batches. For cell-based screening, develop an automated immunocytochemistry workflow using HRP-conjugated SRGAP3 antibodies with computerized image acquisition and analysis parameters. Establish rigorous statistical thresholds for hit identification, including Z-factor calculations and coefficient of variation assessments across replicates. Validate screening hits with orthogonal approaches including functional assays measuring RAC1 activity . Create a tiered validation funnel where primary screens using the HRP-conjugated antibody are followed by more detailed mechanistic studies using unconjugated antibodies in conventional formats.

How should researchers troubleshoot non-specific background when using HRP-conjugated SRGAP3 antibodies in immunohistochemistry?

To address non-specific background with HRP-conjugated SRGAP3 antibodies, implement a systematic troubleshooting approach. First, increase blocking stringency by extending blocking time to 2 hours and using a combination of 5% BSA with 5% normal serum from the same species as the tissue . Quench endogenous peroxidase activity thoroughly using 3% hydrogen peroxide in methanol for 15 minutes prior to antibody application. If background persists, dilute the HRP-conjugated antibody further (try 1:2000 instead of 1:500) and extend primary antibody incubation time at 4°C overnight to favor specific binding kinetics . Add 0.1-0.3% Triton X-100 to all buffers to reduce non-specific hydrophobic interactions. For tissues with high background (like brain), include an avidin/biotin blocking step even when using HRP-conjugated antibodies, as these tissues often have endogenous biotin. Compare staining patterns with known SRGAP3 expression profiles—strong signals in cerebral cortex with minimal staining in colon tissue suggest specific detection . If diffuse background remains, consider switching to a multi-step detection method using unconjugated primary antibody.

How can researchers validate the specificity of SRGAP3 antibody staining patterns in brain tissue?

Validating SRGAP3 antibody specificity in brain tissue requires a multi-level approach. Perform parallel staining using different SRGAP3 antibodies that recognize distinct epitopes (e.g., antibodies targeting AA 201-300 versus AA 900-1000) to confirm consistent localization patterns . Implement peptide competition assays where the immunizing peptide is pre-incubated with the antibody before application to tissue—specific staining should be abolished. Compare staining patterns with mRNA expression data from public databases like Allen Brain Atlas or generate RNA-scope data for SRGAP3 transcript localization. Use tissues from SRGAP3 knockout models as the gold standard negative control, or alternatively, use SRGAP3 siRNA-treated primary neurons to demonstrate reduced staining intensity. Perform dual-labeling with established neuronal markers to confirm that SRGAP3 localization matches its expected distribution in specific neuronal populations. Document staining in multiple brain regions including cerebral cortex (high expression) and regions known to have minimal SRGAP3 expression to demonstrate staining specificity across an expression gradient .

What methodological considerations explain discrepancies in molecular weight observations when detecting SRGAP3 in western blotting?

Discrepancies in SRGAP3 molecular weight detection stem from multiple methodological factors. While the calculated molecular weight is 124 kDa, the observed molecular weight is typically around 125 kDa, with variations possible . Post-translational modifications, particularly phosphorylation of SRGAP3, can increase apparent molecular weight by 2-5 kDa depending on modification sites and extent. Sample preparation conditions affect protein migration—insufficient denaturation in reducing sample buffer or boiling for too short a duration can cause aberrant migration patterns. Gel percentage significantly impacts resolution—use 6-8% acrylamide gels for optimal separation of high molecular weight proteins like SRGAP3 . Buffer systems also influence migration—Tris-glycine versus Tris-tricine systems can yield different apparent molecular weights. When comparing results across studies, note the detection system used—HRP-conjugated primary antibodies may produce slightly different band patterns than two-step detection systems due to potential epitope masking by the HRP conjugation . Alternative splicing of SRGAP3 can generate isoforms with different molecular weights, so brain region-specific variations may represent biological reality rather than technical artifacts .

What are the comparative advantages of using HRP-conjugated versus unconjugated SRGAP3 antibodies in multiplexing experiments?

ParameterHRP-ConjugatedUnconjugated
Protocol lengthShorter (elimination of secondary step)Longer (requires secondary antibody)
Signal amplification potentialLimited to enzymatic amplificationFlexible (can use multiple secondary antibodies)
Spectral compatibilityLimited to HRP-based chromogens/fluorophoresAdaptable to any fluorophore-conjugated secondary
Cross-reactivity riskLower (fewer antibodies in system)Higher (potential secondary antibody cross-reactivity)
Epitope accessibilityPotentially reduced by HRP conjugationMaximized
Sequential labeling compatibilityChallenging (tyramide deposit may block epitopes)Excellent (allows stripping and reprobing)
Cost-effectiveness for large studiesHigher initial cost but fewer reagentsLower primary antibody cost but requires secondaries

What experimental evidence supports the selection of specific SRGAP3 epitope regions for antibody development?

The selection of specific epitope regions for SRGAP3 antibodies is supported by substantial experimental evidence. Antibodies targeting amino acids 201-300 show strong reactivity in multiple applications including ELISA, WB, IF, and IHC, indicating this region contains accessible, immunogenic epitopes that remain available in both native and denatured states . The C-terminal region (AA 956-989) demonstrates excellent specificity for western blotting applications, likely because this region contains sequences that distinguish SRGAP3 from other SRGAP family members . The region spanning amino acids 709-955 has proven particularly successful for generating antibodies with versatile application compatibility, as evidenced by its use in multiple conjugated and unconjugated formats . Structural biology studies suggest these regions represent surface-exposed domains in the folded protein, explaining their accessibility to antibodies in native applications. Epitopes within the 900-1000 amino acid range have demonstrated excellent performance in immunocytochemistry applications, correlating with predicted surface-exposed loops in the protein structure . Importantly, antibodies targeting the N-terminal region show cross-reactivity with multiple species including human, mouse, rat, and even non-mammalian vertebrates like zebrafish, indicating evolutionary conservation of these epitopes .

How do different conjugation strategies affect the performance of SRGAP3 antibodies in various research applications?

Different conjugation strategies significantly impact SRGAP3 antibody performance across applications. HRP conjugation provides exceptional sensitivity in ELISA and chromogenic immunohistochemistry applications due to enzymatic signal amplification, but the bulky enzyme (44 kDa) may restrict epitope access in densely packed tissue regions . Biotin conjugation offers superior versatility through the strong avidin-biotin interaction (Kd ≈ 10^-15 M), enabling multiple detection strategies while maintaining high sensitivity, though endogenous biotin in tissues like brain can increase background . Fluorophore conjugation (FITC) enables direct visualization without enzymatic development steps, ideal for co-localization studies, but provides lower sensitivity than enzyme-based methods without additional amplification . Unconjugated antibodies retain maximum flexibility but require additional detection steps . The table below summarizes application-specific performance:

ConjugationELISAWestern BlotIHCIFMultiplexing Capability
Unconjugated+++ (with optimized secondaries)++++++++++++++
HRP+++++++ (direct chemiluminescence)++++ (tyramide required)+
Biotin+++++++++ (with amplification)+++++
FITC+Not applicable+++ (direct visualization)+++

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.