CSGALNACT2 (Chondroitin Sulfate N-acetylgalactosaminyltransferase-2) antibodies are specialized reagents designed to detect and study the CSGALNACT2 enzyme, a Golgi-resident transferase critical for chondroitin sulfate biosynthesis. These antibodies enable researchers to investigate the enzyme's role in cellular processes, disease mechanisms, and therapeutic targeting. CSGALNACT2 catalyzes the transfer of N-acetylgalactosamine (GalNAc) to glucuronic acid during chondroitin sulfate chain elongation , impacting extracellular matrix structure, cell signaling, and pathogen interactions .
CSGALNACT2 antibodies are primarily polyclonal, produced in rabbits, and validated for diverse research applications. Key technical specifications are summarized below:
These antibodies are strictly for research use (RUO) and not diagnostic/therapeutic purposes .
CSGALNACT2 antibodies were instrumental in demonstrating the enzyme’s interaction with IBDV (Infectious Bursal Disease Virus) VP2 capsid protein. Key findings include:
Golgi Localization: Confocal microscopy confirmed CSGALNACT2’s presence in the Golgi apparatus, colocalizing with VP2 during infection .
Replication Enhancement: Overexpression of CSGALNACT2 increased IBDV replication by 40–60%, while siRNA knockdown reduced viral titers by 70% .
Structural Dependency: Disruption of Golgi integrity (via Brefeldin A) abolished CSGALNACT2’s pro-viral effects, highlighting its structural role .
CSGALNACT2 antibodies revealed its downregulation in ovarian cancer and metastatic tissues:
Expression Loss: IHC showed 3.5-fold lower CSGALNACT2 protein levels in ovarian tumors vs. normal tissues, further reduced in metastases .
Functional Impact:
Immunotherapy Correlation: Low CSGALNACT2 correlates with “immune desert” tumor microenvironments, predicting poor response to anti-PD-L1 therapy .
This antibody targets CSGALNACT2, an enzyme that transfers 1,4-N-acetylgalactosamine (GalNAc) from UDP-GalNAc to the non-reducing end of glucuronic acid (GlcUA). This process is essential for the addition of the first GalNAc to the core tetrasaccharide linker and for the elongation of chondroitin chains.
CSGALNACT2 (Chondroitin Sulfate N-Acetylgalactosaminyltransferase 2) is an enzyme that transfers 1,4-N-acetylgalactosamine (GalNAc) from UDP-GalNAc to the non-reducing end of glucuronic acid (GlcUA). This enzyme plays a critical role in glycosaminoglycan synthesis, being required for both the addition of the first GalNAc to the core tetrasaccharide linker and for elongation of chondroitin chains . CSGALNACT2 functions as a type II transmembrane protein and is predominantly localized in the Golgi apparatus, specifically at the Golgi stack membrane . The protein plays a crucial role in chondroitin sulfate biosynthesis, which has implications in various biological processes including development, cell signaling, and extracellular matrix organization.
CSGALNACT2 antibodies are available in multiple formats optimized for different research applications. These include polyclonal antibodies derived from rabbit hosts that target various epitopes of the CSGALNACT2 protein, as well as monoclonal antibodies like EPR13670 . The availability spans various target regions including amino acids 33-270, 61-110, and 200-229 . Most commercially available antibodies are unconjugated, though some are available with conjugates such as FITC, HRP, or Biotin for specialized applications . The following table summarizes the main types available:
Antibody Type | Host | Target Region | Applications | Reactivity | Catalog Examples |
---|---|---|---|---|---|
Polyclonal | Rabbit | AA 33-270 | ELISA, WB, IHC | Human | ABIN7147640 |
Polyclonal | Rabbit | AA 61-110 | IHC/IF, ELISA | Human, Mouse | STJ92494 |
Polyclonal | Rabbit | AA 200-229 | WB, FACS, IHC(p) | Human | RB23569 |
Monoclonal | Rabbit | Not specified | WB | Human | ab181250 (EPR13670) |
Selecting the appropriate CSGALNACT2 antibody requires careful consideration of several factors based on your experimental design. First, determine which application you need the antibody for—whether it's Western blotting (WB), immunohistochemistry (IHC), ELISA, or flow cytometry (FACS). For instance, if performing Western blot analysis on human cell lines, antibodies validated specifically for WB with human reactivity, such as the rabbit recombinant monoclonal EPR13670, would be appropriate . For immunofluorescence studies, antibodies validated for IF applications are essential .
Second, consider the specific region of CSGALNACT2 you wish to target. If studying the full-length protein, antibodies targeting regions away from functional domains may be preferred, while those investigating specific protein interactions might select antibodies targeting interaction domains. For example, research investigating CSGALNACT2's interaction with viral proteins might benefit from antibodies targeting regions involved in these interactions .
Third, evaluate species reactivity—ensure the antibody recognizes CSGALNACT2 from your experimental species. Most available antibodies target human CSGALNACT2, though some cross-react with mouse or rat variants . Finally, consider clonality: polyclonal antibodies offer broader epitope recognition but potential batch variation, while monoclonal antibodies provide consistency but more restricted epitope binding .
For optimal Western blotting with CSGALNACT2 antibodies, cell lysate preparation is critical. Based on published research protocols, cells should be lysed using a western and immunoprecipitation lysis buffer containing protease inhibitors (e.g., 1 mM PMSF) . For human cell lines such as 293, Jurkat, K562, or Raji, 20 μg of protein lysate per lane is typically sufficient . The predicted band size for CSGALNACT2 is approximately 63 kDa, though post-translational modifications may affect migration patterns .
For immunoblotting, monoclonal antibodies like EPR13670 (ab181250) can be used at a 1/10000 dilution, while polyclonal antibodies may require more concentrated dilutions between 1/1000-1/5000 . Secondary antibody selection should match the host species of the primary antibody—typically anti-rabbit IgG conjugated with peroxidase at 1/1000-1/5000 dilution . To verify specificity, positive controls using cell lines known to express CSGALNACT2 (such as K562 or Jurkat) are recommended, and negative controls can include CSGALNACT2-knockdown samples or competing peptide blocking .
For optimization, titrate your antibody concentration if signal strength is inadequate or background is excessive. Extended blocking steps (1-2 hours) with 3-5% BSA or non-fat milk can help reduce background. When troubleshooting inconsistent results, consider the specific epitope region targeted by your antibody and whether your sample preparation might affect epitope accessibility.
Optimizing immunohistochemistry protocols for CSGALNACT2 detection requires careful attention to fixation, antigen retrieval, and blocking steps. For tissue fixation, 4% paraformaldehyde is commonly used in research protocols . Since CSGALNACT2 is a Golgi-resident protein, membrane permeabilization with 0.1% Triton X-100 is crucial for antibody access to intracellular epitopes .
For antigen retrieval, heat-induced epitope retrieval in citrate buffer (pH 6.0) is generally effective for CSGALNACT2 detection. When blocking, 3% bovine serum albumin (BSA) has been successfully used in published protocols . For primary antibody incubation, polyclonal antibodies targeting amino acids 61-110 can be used at dilutions of 1:100-1:300 for IHC applications .
To validate specificity, co-localization studies with Golgi markers like GOLGA2 can confirm the expected subcellular localization of CSGALNACT2 . The protein should show a characteristic Golgi staining pattern, appearing as perinuclear punctate or crescent-shaped structures. For double immunostaining experiments, selecting primary antibodies from different host species will facilitate discrimination between targets. Additionally, treating samples with Brefeldin A, which disrupts Golgi structure, can serve as a useful negative control by altering the characteristic CSGALNACT2 staining pattern .
Validating CSGALNACT2 antibody specificity is crucial for ensuring experimental reliability. A multi-faceted approach incorporating several validation methods is recommended. First, perform Western blot analysis with positive controls (cell lines with known CSGALNACT2 expression like K562 or Jurkat) and verify the presence of a single band at the expected molecular weight (approximately 63 kDa) . To further confirm specificity, employ RNA interference techniques using siRNA targeting CSGALNACT2, as documented in published protocols . The disappearance or significant reduction of the signal following knockdown strongly supports antibody specificity.
Immunoprecipitation followed by mass spectrometry can provide definitive confirmation that the antibody is capturing CSGALNACT2. For co-immunoprecipitation experiments, researchers have successfully used truncated CSGALNACT2 constructs (p∆CSGALNACT2) with deleted signal peptide and transmembrane regions (amino acids 1-37) to improve solubility . Additionally, peptide competition assays, where pre-incubation of the antibody with the immunizing peptide blocks specific binding, can demonstrate epitope specificity.
For immunolocalization studies, co-localization with established Golgi markers like GOLGA2 should be demonstrated, as CSGALNACT2 is known to localize to the Golgi apparatus . Lastly, utilizing multiple antibodies targeting different epitopes of CSGALNACT2 can provide converging evidence of specificity when they yield consistent results.
CSGALNACT2 antibodies can be powerful tools for investigating the protein's role in viral infection pathways, particularly in the context of infectious bursal disease virus (IBDV) infection. Research has demonstrated that CSGALNACT2 interacts with the viral capsid protein VP2, and this interaction appears to promote viral replication . To study this mechanism, co-immunoprecipitation assays can be performed using anti-CSGALNACT2 antibodies to pull down protein complexes from infected cells, followed by Western blotting for viral proteins like VP2 .
Confocal microscopy utilizing fluorescently labeled CSGALNACT2 antibodies allows visualization of its co-localization with viral proteins during different stages of infection. In published protocols, researchers have co-transfected cells with tagged constructs (FLAG-tagged CSGALNACT2 and HA-tagged VP2) and performed double immunofluorescence using the respective antibodies . Time-course experiments can reveal temporal changes in CSGALNACT2 expression and localization during viral infection.
For functional analyses, researchers can combine CSGALNACT2 antibodies with siRNA-mediated knockdown or overexpression of CSGALNACT2. Previous studies have shown that overexpression of CSGALNACT2 promotes IBDV replication, while knockdown impairs it . To investigate the importance of Golgi integrity in this process, experiments can include Brefeldin A treatment, which disrupts Golgi structure and inhibits CSGALNACT2's enhancing effect on viral replication . Quantitative PCR for viral genes and viral titer assays (TCID50) following manipulation of CSGALNACT2 expression can provide quantitative insights into its role in the viral life cycle.
Studying CSGALNACT2's role in glycosaminoglycan biosynthesis requires sophisticated approaches targeting enzyme activity, substrate specificity, and pathway interactions. In vitro reconstitution assays using soluble forms of CSGALNACT2 (where the cytoplasmic and transmembrane regions are replaced with a signal peptide) have been successfully employed to study its enzymatic function . These assays can measure the transfer of GalNAc to various substrates using radioisotope-labeled UDP-GalNAc as a donor, followed by chromatographic separation and quantification of reaction products.
Antibodies can be used to investigate CSGALNACT2's interactions with other glycosyltransferases in the biosynthetic pathway. Immunoprecipitation with CSGALNACT2 antibodies followed by mass spectrometry can identify associated proteins. Western blotting with antibodies against known glycosyltransferases (such as XT1, B4GALT7, B3GALT6, B3GAT3, and others) can confirm specific interactions .
For investigating the substrate specificity of CSGALNACT2 versus related enzymes like EXTL3, researchers can use antibodies in pulse-chase experiments with metabolic labeling of glycosaminoglycans. This approach can determine the temporal sequence of enzyme recruitment during chain synthesis. Comparative analysis of CSGALNACT2 and EXTL3 activities on different substrate peptides (like BKN and BETA peptides) has revealed that CSGALNACT2 can initiate CS synthesis on all GAG attachment sites, while EXTL3 requires specific features in the core protein to initiate HS synthesis .
For in vivo studies, CSGALNACT2 antibodies can be used for immunohistochemical analysis of tissues from wild-type and CSGALNACT2-knockout models to examine changes in glycosaminoglycan distribution and composition. Combining antibody detection with specific glycosaminoglycan staining techniques provides comprehensive insights into CSGALNACT2's role in tissue-specific glycosaminoglycan synthesis.
CSGALNACT2 antibodies offer valuable tools for investigating chondroitin sulfate modifications in cancer, particularly in relation to oncofetal chondroitin sulfate (ofCS). Recent research has identified ofCS as a potential cancer-specific target, with antibody-based approaches showing promise for tumor-agnostic cancer therapy . For researchers investigating this area, CSGALNACT2 antibodies can be employed to evaluate enzyme expression levels across different tumor types using tissue microarrays and immunohistochemistry. This can establish correlations between CSGALNACT2 expression and clinical parameters such as tumor grade, stage, and patient outcomes.
To investigate the functional significance of CSGALNACT2 in cancer progression, researchers can combine antibody-based protein detection with genetic manipulation approaches. For example, CRISPR-Cas9 knockout or siRNA knockdown of CSGALNACT2 in cancer cell lines, followed by assessment of chondroitin sulfate synthesis, cell proliferation, migration, and invasion assays can reveal its role in malignant phenotypes. In published research, antibody fragments targeting ofCS have demonstrated tumor specificity both in vitro and in vivo, accumulating in xenograft tumors after systemic administration .
For translational applications, CSGALNACT2 antibodies can be used to evaluate the enzyme as a potential therapeutic target or biomarker. Co-localization studies with other glycosyltransferases involved in chondroitin sulfate modification can reveal pathway alterations in cancer cells. Additionally, antibodies can be used to monitor changes in CSGALNACT2 expression and localization in response to therapeutic interventions, providing insights into treatment mechanisms and potential resistance pathways.
Researchers working with CSGALNACT2 antibodies frequently encounter several challenges that require systematic troubleshooting. One common issue is weak or absent signal in Western blot applications. This may be addressed by optimizing protein extraction protocols specific for membrane-associated Golgi proteins—utilizing lysis buffers containing 0.5-1% nonionic detergents (Triton X-100 or NP-40) helps solubilize CSGALNACT2 effectively . For samples with low CSGALNACT2 expression, enrichment techniques such as immunoprecipitation prior to Western blotting or using more sensitive detection systems (enhanced chemiluminescence plus or fluorescent secondary antibodies) can improve detection.
Another challenge is non-specific binding, which manifests as multiple bands in Western blots or diffuse staining in immunohistochemistry. This can be mitigated by more stringent blocking (extending blocking time to 1-2 hours with 5% BSA) and implementing more thorough washing steps with increased salt concentration in PBST (up to 500 mM NaCl) . Titrating primary antibody concentrations can also help identify the optimal dilution that maximizes specific binding while minimizing background.
For immunolocalization studies, the perinuclear Golgi localization of CSGALNACT2 can sometimes be difficult to distinguish from other subcellular structures. Researchers should include co-staining with established Golgi markers like GOLGA2 to confirm proper localization . When working with tissue samples, inconsistent fixation can lead to variable epitope accessibility. Standardizing fixation protocols and implementing appropriate antigen retrieval methods (heat-induced epitope retrieval with citrate buffer pH 6.0) can improve consistency across experiments.
Ensuring reproducibility when working with CSGALNACT2 antibodies requires attention to several key factors. First, implement detailed record-keeping of antibody information including catalog number, lot number, concentration, storage conditions, and freeze-thaw cycles. Different lots of polyclonal antibodies may exhibit variation in epitope recognition, so testing new lots against previous ones is advisable . When possible, utilize monoclonal antibodies like EPR13670 for applications requiring highest consistency .
Second, standardize sample preparation protocols. For cell lines, maintain consistent passage numbers, confluence levels (ideally 80-90%), and lysis conditions . Document and control variables that might affect CSGALNACT2 expression or localization, such as cell stress conditions or treatments that disrupt Golgi structure (like Brefeldin A) .
Third, implement quantitative approaches in data analysis. For Western blots, use densitometry with appropriate normalization to housekeeping proteins. For immunofluorescence, employ quantitative image analysis metrics like colocalization coefficients when assessing CSGALNACT2 distribution relative to organelle markers. The following approach is recommended for validating reproducibility:
Run internal controls consistently across experiments (positive controls like K562 or Jurkat cells, negative controls like knockdown samples)
Perform technical replicates (minimum of three) within each experiment
Conduct biological replicates across different days and sample preparations
Use statistical analyses appropriate for your experimental design to assess variability
Finally, consider antibody validation scoring systems to objectively evaluate reliability. Multiple antibodies targeting different CSGALNACT2 epitopes can be used in parallel to confirm findings are antibody-independent. For critical experiments, orthogonal methods not relying on antibodies (such as mass spectrometry or RNA analysis) provide complementary validation.
Co-localization studies examining CSGALNACT2 alongside other Golgi or glycosylation machinery proteins require careful experimental design and interpretation. First, when selecting antibody combinations, choose primary antibodies raised in different host species to enable simultaneous detection without cross-reactivity. For instance, rabbit anti-CSGALNACT2 can be paired with mouse antibodies against other Golgi proteins like GOLGA2 . When this is not possible, consider sequential staining protocols with intermediate blocking steps using excess unconjugated secondary antibodies.
Third, employ high-resolution imaging techniques appropriate for Golgi studies. Standard confocal microscopy provides good resolution for general co-localization, but super-resolution approaches like structured illumination microscopy (SIM) or stimulated emission depletion (STED) microscopy can reveal subcompartmental distributions within the Golgi apparatus that might otherwise be missed. For quantitative co-localization analysis, utilize appropriate coefficients such as Pearson's correlation coefficient, Manders' overlap coefficient, or object-based co-localization metrics.
Fourth, include appropriate controls to validate the specificity of observed co-localization patterns:
Single-stained samples to assess bleed-through between fluorescence channels
siRNA knockdown of CSGALNACT2 to confirm staining specificity
Brefeldin A treatment to disrupt Golgi organization as a functional control
Co-localization with markers of different Golgi subcompartments (cis, medial, trans) to precisely map CSGALNACT2 distribution
Finally, when interpreting results, remember that optical co-localization does not necessarily indicate molecular interaction. Complementary approaches such as proximity ligation assays or FRET (Fluorescence Resonance Energy Transfer) can provide evidence of direct protein-protein interactions within the spatial resolution limits of light microscopy.
CSGALNACT2 antibodies can be strategically implemented in high-throughput screening (HTS) platforms for drug discovery, particularly for compounds targeting glycosaminoglycan biosynthesis pathways. Cell-based HTS assays utilizing CSGALNACT2 antibodies can be developed using immunofluorescence or ELISA-based detection methods in 384-well or 1536-well formats. For immunofluorescence-based screening, automated image acquisition and analysis systems can quantify changes in CSGALNACT2 expression, localization, or post-translational modifications in response to compound libraries.
A particularly promising approach leverages the finding that CSGALNACT2 promotes virus replication through interaction with viral proteins . Researchers can develop cell-based assays where CSGALNACT2-viral protein interactions are monitored using techniques such as split-luciferase complementation or FRET-based sensors. Compounds disrupting these interactions could be identified as potential antiviral candidates. Alternatively, screens could target compounds that modulate CSGALNACT2 enzymatic activity, potentially affecting glycosaminoglycan biosynthesis in disease states.
For screening specificity, researchers should implement counter-screens with related glycosyltransferases such as CSGALNACT1 or EXTL3 to identify compounds with selective activity against CSGALNACT2 . Confirmation assays following primary screening should include direct enzyme activity measurements, as well as cellular glycosaminoglycan composition analysis using liquid chromatography-mass spectrometry. The most promising compounds can then be validated in more complex cellular models, including 3D organoids or primary cell cultures, using CSGALNACT2 antibodies to confirm target engagement and pathway modulation.
Studying glycosaminoglycan biosynthesis in three-dimensional (3D) cell culture models and organoids presents unique challenges that require specific considerations when using CSGALNACT2 antibodies. First, antibody penetration into 3D structures is more limited than in monolayer cultures. Researchers should optimize fixation and permeabilization protocols—extending incubation times (24-48 hours for antibody penetration) and using more effective permeabilization agents like 0.2-0.5% Triton X-100 or 0.1% saponin with 0.2% gelatin can improve access to internal structures. Additionally, reducing the size of organoids through sectioning (vibratome sections of 100-200 μm) or using clearing techniques (CLARITY, CUBIC, or iDISCO) can dramatically improve antibody penetration and imaging quality.
Second, when interpreting CSGALNACT2 localization in 3D models, researchers must consider that Golgi morphology and organization may differ significantly from 2D cultures. The Golgi apparatus in 3D cultures often shows more physiologically relevant organization that relates to cellular polarization within the model. Therefore, co-staining with multiple Golgi markers becomes even more important to accurately interpret CSGALNACT2 distribution. For quantitative analysis, confocal z-stacks spanning the entire structure should be acquired, and 3D rendering software should be used to visualize the complete distribution pattern.
Third, accounting for the extracellular matrix (ECM) surrounding cells in 3D models is crucial, as the ECM contains glycosaminoglycans that are the products of CSGALNACT2 activity. To distinguish between cellular CSGALNACT2 and its extracellular products, researchers can implement dual staining approaches—using antibodies against CSGALNACT2 together with specific glycosaminoglycan detection methods such as anti-chondroitin sulfate antibodies or specific glycosaminoglycan-binding proteins. Time-course experiments tracking newly synthesized glycosaminoglycans can provide insights into the dynamics of synthesis and deposition in the 3D microenvironment.
Investigating the cross-talk between glycosaminoglycan biosynthesis and other post-translational modifications (PTMs) requires sophisticated experimental approaches utilizing CSGALNACT2 antibodies. Multiplexed immunofluorescence combining antibodies against CSGALNACT2 with those targeting proteins involved in other PTM pathways (such as protein kinases, ubiquitin ligases, or other glycosylation enzymes) can reveal spatial relationships between these different modification machineries. Super-resolution microscopy techniques are particularly valuable for resolving potentially subtle co-localizations within Golgi subcompartments.
To examine functional relationships, researchers can employ perturbation approaches followed by immunoblotting with CSGALNACT2 antibodies. For instance, inhibiting specific kinase pathways (using small molecule inhibitors or CRISPR-mediated knockout) and subsequently analyzing changes in CSGALNACT2 expression, localization, or post-translational state can reveal regulatory connections. Phosphorylation site-specific antibodies can be developed to monitor CSGALNACT2 phosphorylation status under different conditions, providing insights into its regulation.
For comprehensive PTM profiling, immunoprecipitation with CSGALNACT2 antibodies followed by mass spectrometry analysis can identify multiple modifications on the protein itself. Researchers can implement SILAC (Stable Isotope Labeling with Amino acids in Cell culture) or TMT (Tandem Mass Tag) labeling to quantitatively compare CSGALNACT2 PTMs across different cellular conditions. Alternatively, proximity labeling approaches like BioID or APEX2 can be used by creating fusion proteins with CSGALNACT2, enabling identification of proteins in its vicinity that might be involved in cross-regulatory relationships.
To specifically address the interplay between different glycosylation pathways, researchers can manipulate CSGALNACT2 expression or activity and examine effects on N-glycosylation, O-glycosylation, or other glycosaminoglycan types using lectin arrays or mass spectrometry-based glycomics. Such studies may reveal compensatory mechanisms or competitive relationships between different glycosylation pathways that share precursor molecules or regulatory mechanisms.