SDCBP Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction

SDCBP (Syndecan Binding Protein), also known as Syntenin-1, is a scaffolding protein implicated in cell adhesion, migration, intracellular trafficking, and cancer progression. SDCBP monoclonal antibodies are laboratory-generated immunoreagents designed to specifically detect and study this protein. These antibodies are critical for research applications such as Western blotting (WB), immunohistochemistry (IHC), and therapeutic development.

Hybridoma-Based Production

  • Immunogen: Recombinant human SDCBP protein fragments (e.g., residues 2–298 or 1–100) are used to immunize mice or rabbits .

  • Hybridoma Creation: B cells from immunized animals are fused with myeloma cells to generate antibody-producing hybridomas .

  • Purification: Antibodies are purified via protein G affinity chromatography, achieving >95% purity .

Recombinant Production

  • In Vitro Cloning: Antibody genes are cloned into plasmid vectors and expressed in host cells (e.g., HEK293) .

  • Epitope Specificity: Most target linear epitopes within the N-terminal region (e.g., residues 1–100) .

ParameterHybridoma Antibody (e.g., Cusabio CSB-MA020893A0m)Recombinant Antibody (e.g., Cusabio CSB-RA248860A0HU)
Host SpeciesMouseRabbit
ReactivityHuman, Mouse, RatHuman
ApplicationsWB, ELISAWB, IF, FC
Clone2C12 (IgG2b) N/A (Recombinant)
Purity>95%>95%

Research Applications and Validation

SDCBP monoclonal antibodies are widely used in both basic and translational research:

Key Applications

  • Western Blotting: Detects SDCBP at ~32–39 kDa in human cell lines (e.g., HepG2, HeLa) .

  • Immunofluorescence (IF): Localizes SDCBP to cytoplasm, membrane, and endosomes .

  • Therapeutic Development: Anti-SDCBP IgG4 reduces tumor volume in ovarian cancer models .

Validation Data

  • Knockdown Validation: siRNA-mediated SDCBP silencing in PNT2 cells reduced antibody signal by 4–16-fold .

  • Cross-Reactivity: Commercial polyclonal antibodies show cross-reactivity with unrelated proteins (e.g., 130 kDa species in 22RV1 cells), whereas monoclonal antibodies (e.g., Synt-2C6) exhibit higher specificity .

Role in Cancer

  • Glioma Progression: SDCBP expression correlates with tumor grade (r = 0.661, P < 0.001) and SDC1 levels (r = 0.628, P = 0.001) in gliomas .

  • Ovarian Cancer: An α-SDCBP IgG4 reduced tumor volume by 50% in TOV21G (clear cell carcinoma) and OVCAR3 (high-grade serous) mouse models .

SDC1 vs. SDCBP Expression in Gliomas
SDC1 StatusSDCBP+
Positive (n=23)20
Negative (n=9)2
Data from 32 glioma tissues; Spearman’s correlation = 0.628 (P=0.001)

Challenges and Considerations

  • Cross-Reactivity: Polyclonal antibodies may detect non-target proteins (e.g., 130 kDa species) .

  • Storage: Stable at -20°C; avoid repeated freeze-thaw cycles .

  • Species Limitations: Most antibodies are validated for human samples; cross-reactivity with primates requires pilot testing .

Future Directions

SDCBP monoclonal antibodies hold promise for:

  1. Biomarker Development: Leveraging SDCBP’s overexpression in gliomas and ovarian cancers .

  2. Targeted Therapy: Optimizing antibody-drug conjugates (ADCs) for SDCBP+ tumors.

  3. Mechanistic Studies: Elucidating SDCBP’s role in Wnt/TGF-β signaling and cytoskeletal dynamics .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

The SDCBP monoclonal antibody is produced through a meticulous process. Mice are immunized with recombinant human SDCBP protein (amino acids 2-298), and B cells are isolated from their spleens. These B cells are then fused with myeloma cells to create hybridomas. Through careful screening, the hybridoma cell line that produces the SDCBP antibody is selected and cultured in the mouse abdominal cavity. The purified SDCBP monoclonal antibody is subsequently obtained from the mouse ascites using protein G affinity chromatography, guaranteeing a purity exceeding 95%. This unconjugated IgG2b antibody exhibits reactivity across three species: human, mouse, and rat, making it suitable for applications such as ELISA and Western blotting.

SDCBP, also recognized as Syntenin-1, is a multifaceted protein involved in a diverse array of cellular processes, including cell adhesion, migration, and signaling. Acting as a scaffolding protein, SDCBP interacts with a range of membrane and cytoplasmic proteins, such as syndecans, integrins, and signaling molecules, to form multi-protein complexes. These complexes play a crucial role in the regulation of cell-cell and cell-matrix interactions, as well as in modulating intracellular signaling pathways, including those governed by the Wnt and TGF-β signaling pathways. SDCBP has also been implicated in the regulation of endocytosis, exocytosis, and vesicular trafficking.

Form
Liquid
Lead Time
Typically, we are able to dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the mode of purchase and location. For specific delivery timeframes, please consult your local distributors.
Synonyms
MDA-9 antibody; MDA9 antibody; Melanoma differentiation-associated protein 9 antibody; Pro-TGF-alpha cytoplasmic domain-interacting protein 18 antibody; Scaffold protein Pbp1 antibody; SDCB1_HUMAN antibody; SDCBP antibody; ST1 antibody; SYCL antibody; Syndecan binding protein (syntenin) antibody; Syndecan binding protein 1 antibody; Syndecan-binding protein 1 antibody; Syntenin 1 antibody; Syntenin-1 antibody; TACIP18 antibody
Target Names
SDCBP
Uniprot No.

Target Background

Function
Syntenin-1, a multifunctional adapter protein, plays a critical role in a wide range of cellular functions, including the trafficking of transmembrane proteins, neuro and immunomodulation, exosome biogenesis, and tumorigenesis. It acts as a positive regulator of TGFβ1-mediated SMAD2/3 activation, driving TGFβ1-induced epithelial-to-mesenchymal transition (EMT) and cell migration in various cell types. Syntenin-1 enhances TGFβ1 signaling by promoting cell-surface expression of TGFβR1, preventing the interaction between TGFβR1 and CAV1 and subsequent CAV1-dependent internalization and degradation of TGFβR1. In concert with SDC1/4 and PDCD6IP, Syntenin-1 regulates exosome biogenesis. Further, it regulates migration, growth, proliferation, and cell cycle progression in various cancer types. Within adherens junctions, Syntenin-1 may function to link syndecans to cytoskeletal proteins or signaling components. It appears to couple the transcription factor SOX4 to the IL-5 receptor (IL5RA). Additionally, Syntenin-1 may contribute to vesicular trafficking and is believed to be essential for the targeting of TGFA to the cell surface during the early secretory pathway.
Gene References Into Functions
  1. Investigated the role of melanoma differentiation-associated protein-9 (MDA-9)/Syntenin in autophagy of anoikis-resistant glioma stem cells. PMID: 29760085
  2. Examined how Frizzled 7 and phosphatidylinositol 4,5-diphosphate binding by the syntenin PDZ2 domain supports Frizzled 7 trafficking and signaling. PMID: 27386966
  3. Demonstrated that syntenin mediates SRC function in exosomal cell-to-cell communication. PMID: 29109268
  4. These findings indicate that syntenin promotes VEGF signaling and, through its PDZ-dependent interaction with ephrin-B2, enhances VEGF-mediated VEGFR2 endocytosis, subsequent downstream signaling, and angiogenesis in endothelial cells. PMID: 28418925
  5. Syntenin-1 promotes invasion and progression of squamous cell carcinomas of the head and neck. PMID: 27811365
  6. MDA-9 upregulated active levels of known modulators of epithelial mesenchymal transformation, the small GTPases RhoA and Cdc42, via TGFbeta1, promoting lung metastasis of breast cancer cells. PMID: 27863394
  7. Identified a unique function of MDA-9 as a facilitator and determinant of glioma stemness and survival. PMID: 27472461
  8. SDCBP might be a valuable marker for identifying Triple-negative breast cancer cases that are suitable for dasatinib therapy. PMID: 28141839
  9. ACTB, CDKN1B, GAPDH, GRB2, RHOA, and SDCBP are potent reference genes in neuroendocrine tumors of the lung. PMID: 27802291
  10. This research suggests that miR-216b acts as a regulator of SDCBP expression in breast cancer, potentially offering a target for developing novel therapies to effectively treat this disease. PMID: 27720715
  11. Data indicate that patients with high MDA-9/Syntenin and high Slug expressions were associated with poorer overall survival compared to those with low expression in lung adenocarcinomas. PMID: 26561205
  12. Research suggests that the syntenin/SDCBP PDZ domains 1 and 2 recognize a broad range of peptide ligands, exhibiting preferences for nectin-1, hydrophobic amino acid motifs, and cryptic internal ligands/peptide fragments. PMID: 26787460
  13. These findings demonstrate that syntenin may function as a significant positive regulator of TGF-β signaling by controlling caveolin-1-mediated internalization of TbRI, thereby providing a novel function for syntenin linked to cancer progression. PMID: 25893292
  14. To predict mda-9's association with extracellular matrix organization. PMID: 26093898
  15. MDA-9, co-expressed with GRP78, is identified as a melanoma protein associated with lymph node metastasis. Investigating the interaction of MDA-9 and GRP78 in contributing to melanoma metastasis and disease progression could reveal promising avenues for targeted therapies. PMID: 25480418
  16. Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. PMID: 24637612
  17. High MDA-9 expression is linked to glioma. PMID: 24305713
  18. ALCAM stably interacts with actin by binding to syntenin-1 and ezrin. PMID: 24662291
  19. These findings indicate that MDA-9/Syntenin might provide a compelling target for developing strategies for detection, monitoring, and therapeutic intervention in Urothelial cell carcinoma. PMID: 23873690
  20. Results suggest that SDCBP plays a significant role in tumor growth of ER-negative breast cancers. PMID: 23533663
  21. Findings establish RKIP as an inhibitor of MDA-9-dependent melanoma metastasis. PMID: 23066033
  22. Our studies unveil an unexpected cell nonautonomous function of MDA-9/syntenin in the context of angiogenesis, which may directly contribute to its metastasis-promoting properties. PMID: 23233738
  23. Results demonstrate that Mda-9/syntenin overexpression can activate FAK-JNK and FAK-Akt signaling, enhancing the migration capacity of human brain glioma cells. PMID: 22938480
  24. Data show that syntenin-1 is recruited to the plasma membrane during HIV-1 attachment. PMID: 22535526
  25. The study highlights the key role of syntenin-1 in generating functional asymmetry in T cells, providing a novel mechanistic link between receptor activation and actin polymerization and accumulation in response to extracellular stimulation. PMID: 22349701
  26. Our data demonstrate that the Sox4 C-terminal domain regulates polyubiquitin-independent proteasomal degradation of Sox4, which can be modulated by interaction with syntenin. PMID: 21986941
  27. MDA-9/syntenin functions as a positive regulator of melanoma progression and metastasis through interactions with c-Src, promoting the formation of an active FAK/c-Src signaling complex, leading to NF-κB and matrix metalloproteinase activation. Review. PMID: 22201728
  28. Mda-9/syntenin is involved in uveal melanoma progression. PMID: 22267972
  29. A ubiquitin-dependent pathway involving syntenin-1 is regulated by Ulk1. PMID: 21949238
  30. Mda-9/syntenin, a positive regulator of cancer metastasis, regulates the activation of Akt (also known as protein kinase B) by facilitating ILK adaptor function during adhesion to type I collagen (COL-I) in human breast cancer cells. PMID: 21828040
  31. Research showed that overexpression of wild-type MDA-9/syntenin enhances the formation of filopodia, whereas MDA-9/syntenin lacking the PDZ domain inhibits the formation of filopodia. PMID: 21359963
  32. ST1 were up-regulated with the malignancy of prostate cancer cell lines and have their potential as serum biomarkers for indicating the developmental stage of prostate cancer. PMID: 20233700
  33. The large/zonula occludens-1 domains of MDA-9 represent a promising potential therapeutic target for preventing cancer progression and metastatic spread. PMID: 20228839
  34. Data indicate that the functional properties of syntenin are a result of independent interactions with target peptides. PMID: 12679023
  35. Melanoma metastasis is associated with increased expression of the syntenin gene, which may participate in signal transduction and cell adhesion via the multifunctional protein-binding properties of its tandem PDZ domains. PMID: 15254681
  36. eIF5A may be a regulator of p53, and syntenin might regulate p53 by balancing the regulation of eIF5A signaling to p53 for apoptosis. PMID: 15371445
  37. This review discusses the identification, structure, and function of mda-9/syntenin and delineates future studies to address its role in regulating key physiological and pathological processes. PMID: 15518882
  38. This study provides the first direct link between mda-9/syntenin expression and tumor cell dissemination in vivo and indicates that mda-9/syntenin expression activates specific signal transduction pathways. PMID: 16322237
  39. Collectively, these results suggest that downregulation of syntenin by RNA interference could provide a means of inhibiting tumor invasion and possibly metastasis in different cancers, and point to syntenin as a potential cancer biomarker and drug target. PMID: 17451681
  40. Syntenin binding to Delta1 plays a dual role in promoting intercellular adhesion and regulating Notch signaling. PMID: 17666427
  41. This research suggests that syntenin is a physiological suppressor of TRAF6 and plays an inhibitory role in IL-1R- and TLR4-mediated NF-κB activation pathways. PMID: 18234474
  42. Syntenin stimulates c-jun phosphorylation and modulates Frizzled 7 signaling, particularly the PKCalpha/CDC42 noncanonical Wnt signaling cascade. PMID: 18256285
  43. Data are compatible with a model wherein interaction of MDA-9/syntenin with c-Src promotes the formation of an active FAK/c-Src signaling complex, leading to enhanced tumor cell invasion and metastatic spread. PMID: 18832467
  44. Syntenin interacts with the aminoacyl tRNA synthetase complex in a lysyl-tRNA synthetase-dependent manner. PMID: 18839981
  45. Syntenin-1 serves as one of the IgA-inducing factors for B cells. PMID: 19592421
  46. Syntenin forms complexes with multiple IL-5Ralpha chains. PMID: 19654410

Show More

Hide All

Database Links

HGNC: 10662

OMIM: 602217

KEGG: hsa:6386

STRING: 9606.ENSP00000260130

UniGene: Hs.200804

Subcellular Location
Cell junction, focal adhesion. Cell junction, adherens junction. Cell membrane; Peripheral membrane protein. Endoplasmic reticulum membrane; Peripheral membrane protein. Nucleus. Melanosome. Cytoplasm, cytosol. Cytoplasm, cytoskeleton. Secreted, extracellular exosome. Membrane raft.
Tissue Specificity
Expressed in lung cancers, including adenocarcinoma, squamous cell carcinoma and small-cell carcinoma (at protein level). Widely expressed. Expressed in fetal kidney, liver, lung and brain. In adult highest expression in heart and placenta.

Q&A

What is SDCBP and why is it important in cancer research?

SDCBP (Syndecan Binding Protein, also known as syntenin) is a scaffold protein containing two PDZ domains that has been identified as a molecule binding to the cytoplasmic domain of syndecans . SDCBP is particularly important in cancer research because it serves as an important mediator of invasion in several cancers including glioma . Immunohistochemical analysis has revealed that SDCBP expression is positively related to the malignant level of glioma (rs=0.661, P<0.001), with high-grade gliomas showing the strongest staining intensity and the highest number of positive-staining cells . To effectively study SDCBP in cancer contexts, researchers should employ multiple detection methods including immunohistochemistry, western blotting, and functional assays to establish correlation between SDCBP expression and tumor phenotypes.

How do SDCBP monoclonal antibodies differ from other research antibodies?

SDCBP monoclonal antibodies are highly specific research tools that recognize distinct epitopes of the SDCBP protein. Unlike polyclonal antibodies that recognize multiple epitopes, monoclonal antibodies offer greater specificity and reproducibility in experimental settings. When selecting SDCBP monoclonal antibodies for research, it's crucial to validate their specificity through positive and negative controls, particularly in cell lines with known SDCBP expression patterns . Western blotting validation should confirm a band at the expected molecular weight (approximately 32 kDa for SDCBP), while immunocytochemistry should reveal primarily cytoplasmic localization as observed in glioma tissues .

What is the relationship between SDC1 and SDCBP in experimental systems?

The relationship between Syndecan-1 (SDC1) and SDCBP has been experimentally validated in several studies. Spearman correlation analysis shows a positive relationship between protein levels of SDC1 and SDCBP in glioma tissues (rs=0.628, P=0.001) . In U251 glioma cells, manipulation of SDC1 expression directly affects SDCBP levels - SDC1 overexpression significantly increases SDCBP protein levels (SDC1-OE: 0.284±0.044; control: 0.153±0.013; vector: 0.146±0.036; P=0.004), while SDC1 knockdown decreases SDCBP expression (siSDC1: 0.077±0.019; control: 0.201±0.015; siNC: 0.193±0.044; P=0.004) . For researchers investigating this relationship, it's advisable to design experiments that manipulate one protein while monitoring effects on the other, using both overexpression and knockdown approaches to establish causality.

How should researchers optimize immunohistochemistry protocols for SDCBP detection in tissue samples?

When optimizing immunohistochemistry protocols for SDCBP detection in tissue samples, researchers should be aware that SDCBP primarily localizes in the cytoplasm, as demonstrated in glioma tissue analysis . For optimal results, consider the following methodological approaches:

  • Fixation and antigen retrieval: Use 10% neutral-buffered formalin for tissue fixation followed by heat-induced epitope retrieval in citrate buffer (pH 6.0).

  • Antibody concentration: Titrate SDCBP monoclonal antibodies starting at 1:100-1:500 dilutions to determine optimal signal-to-noise ratio.

  • Detection systems: Employ sensitive detection systems such as polymer-based detection rather than traditional avidin-biotin methods.

  • Controls: Include positive controls (high-grade glioma tissues) and negative controls (non-tumorous brain tissues) as SDCBP expression is significantly higher in high-grade gliomas compared to non-tumorous brain tissues .

  • Scoring system: Implement a standardized scoring system similar to that used in published studies, where staining intensity is graded as negative (-), weak (+), moderate (++), or strong (+++) .

What are the critical parameters for using SDCBP antibodies in Western blotting?

When using SDCBP monoclonal antibodies for Western blotting, several critical parameters must be optimized:

  • Sample preparation: Extract proteins using RIPA buffer supplemented with protease inhibitors to prevent SDCBP degradation.

  • Protein loading: Load 20-40 μg of total protein per lane for optimal detection of SDCBP.

  • Gel percentage: Use 10-12% SDS-PAGE gels for optimal resolution of SDCBP (~32 kDa).

  • Transfer conditions: Perform wet transfer at 100V for 1 hour using PVDF membranes for better protein retention.

  • Blocking: Block membranes with 5% non-fat dry milk in TBST for 1 hour at room temperature.

  • Primary antibody incubation: Dilute SDCBP monoclonal antibody 1:1000-1:2000 in blocking buffer and incubate overnight at 4°C.

  • Normalization: Use β-actin or GAPDH as loading controls for accurate quantification of SDCBP expression.

  • Quantification: Perform densitometric analysis of Western blot bands using software like ImageJ to obtain relative expression levels, similar to the quantification approach used in the U251 cell study (SDC1-OE: 0.284±0.044; control: 0.153±0.013) .

How can researchers effectively use SDCBP antibodies in cell migration studies?

To effectively use SDCBP antibodies in cell migration studies, researchers should implement a comprehensive experimental design:

  • Establish cellular models: Use cell lines with manipulated SDCBP expression levels through overexpression or knockdown approaches, as demonstrated with SDC1 in U251 glioma cells .

  • Functional migration assays: Employ both Transwell assay and scratch-wound healing assay to comprehensively assess cell migration capability .

  • Antibody application: Use SDCBP monoclonal antibodies to:

    • Confirm SDCBP expression levels via Western blotting before migration assays

    • Visualize SDCBP localization during migration using immunofluorescence

    • Potentially block SDCBP function using neutralizing antibodies

  • Downstream signaling: Investigate associated pathways affected by SDCBP expression, particularly Rac1 activity, STAT3 phosphorylation, and MMP2/MMP9 expression, which have been shown to be regulated by SDC1/SDCBP interaction .

  • Quantification: Analyze migration rate by measuring the distance traveled by cells in scratch assays or counting cells that passed through the Transwell membrane, normalizing to control conditions.

How can researchers investigate the molecular mechanisms of SDCBP-mediated glioma invasion?

Investigating the molecular mechanisms of SDCBP-mediated glioma invasion requires a multi-faceted approach:

  • SDCBP protein interaction network analysis:

    • Perform co-immunoprecipitation using SDCBP monoclonal antibodies to identify binding partners

    • Validate interactions with known partners like SDC1, which shows a positive correlation with SDCBP expression in glioma tissues (rs=0.628, P=0.001)

    • Use proximity ligation assays to confirm interactions in situ

  • Signaling pathway investigation:

    • Measure Rac1 activity using SDCBP antibodies in pull-down assays, as Rac1 activation has been linked to SDC1/SDCBP function in glioma migration

    • Assess STAT3 phosphorylation levels in relation to SDCBP expression

    • Quantify MMP2 and MMP9 expression and activity using gelatin zymography

  • In vitro 3D invasion models:

    • Use SDCBP antibodies for immunofluorescence visualization in 3D spheroid invasion assays

    • Correlate SDCBP localization with invasion fronts in 3D cultures

  • In vivo models:

    • Employ SDCBP antibodies for immunohistochemical analysis of tumor invasion in orthotopic glioma xenograft models

    • Correlate SDCBP staining with invasive tumor margins

  • Clinical correlation:

    • Analyze SDCBP expression patterns in relation to patient outcomes and invasion parameters in human glioma samples of varying grades

What role does SDCBP play in the SDC1-Rac1-STAT3 signaling axis in glioma?

SDCBP serves as a critical mediator in the SDC1-Rac1-STAT3 signaling axis in glioma progression:

  • Regulatory relationship:

    • SDC1 overexpression upregulates SDCBP expression in U251 glioma cells, while SDC1 knockdown decreases SDCBP levels

    • This regulatory relationship suggests SDCBP acts downstream of SDC1 in signaling cascades

  • Rac1 activation mechanism:

    • SDC1 overexpression significantly increases Rac1 activity, while SDC1 knockdown decreases it

    • SDCBP likely serves as an adaptor that links SDC1 to Rac1 activation components

    • Researchers should examine SDCBP's PDZ domains for interaction with guanine nucleotide exchange factors that activate Rac1

  • STAT3 phosphorylation:

    • SDC1 manipulation affects STAT3 phosphorylation levels, with SDC1 overexpression increasing and knockdown decreasing STAT3 phosphorylation

    • The mechanism likely involves SDCBP-mediated signaling between Rac1 and STAT3

  • MMP regulation:

    • The expression of MMP2 and MMP9 is significantly affected by SDC1 levels

    • SDCBP may facilitate transcriptional regulation of these invasion-promoting enzymes through the STAT3 pathway

  • Experimental approach:

    • Use SDCBP monoclonal antibodies for co-immunoprecipitation studies to identify direct binding partners in this signaling axis

    • Apply proximity ligation assays to visualize SDCBP-Rac1 interactions in situ

    • Perform chromatin immunoprecipitation to investigate STAT3 binding to MMP promoters in relation to SDCBP expression

How can SDCBP monoclonal antibodies be utilized in developing therapeutic strategies for glioma?

SDCBP monoclonal antibodies could contribute to developing therapeutic strategies for glioma through several research approaches:

  • Target validation studies:

    • Use SDCBP antibodies to comprehensively map expression patterns across glioma grades, confirming its positive correlation with malignancy (rs=0.661, P<0.001)

    • Compare SDCBP expression in treatment-responsive versus resistant tumors

  • Functional blocking studies:

    • Develop and test function-blocking SDCBP antibodies that interfere with its PDZ domain interactions

    • Assess effects on glioma cell migration, invasion, and signaling pathways in vitro

  • Antibody engineering approaches:

    • Consider engineering SDCBP-targeted antibodies with brain-penetrating capabilities similar to approaches used for other therapeutic antibodies

    • Explore transferrin conjugation, which has been shown not to alter binding to brain proteins

  • Combination therapy research:

    • Investigate the effects of combining SDCBP-targeting approaches with conventional glioma treatments

    • Use SDCBP antibodies to monitor treatment-induced changes in signaling pathways

  • Blood-brain barrier considerations:

    • Address BBB penetration challenges that have limited other therapeutic antibodies, as CNS exposure for circulating biologics is typically limited to 0.1 to 0.4% of corresponding serum concentrations

    • Explore antibody fragment approaches, as smaller fragments may have better BBB penetration than full antibodies

What are common challenges in detecting SDCBP in brain tissue samples and how can they be overcome?

Researchers commonly encounter several challenges when detecting SDCBP in brain tissue samples:

  • Low signal-to-noise ratio:

    • Challenge: SDCBP shows weak signal in non-tumorous brain tissues, where positive-staining cells are scattered

    • Solution: Optimize antigen retrieval conditions using citrate or EDTA buffers at various pH levels; employ signal amplification systems like tyramide signal amplification

  • Variable expression across samples:

    • Challenge: SDCBP expression varies significantly between high-grade gliomas, low-grade gliomas, and non-tumorous tissues

    • Solution: Incorporate appropriate positive controls (high-grade glioma) and negative controls (non-tumorous brain tissue) in each experiment; standardize tissue processing and staining procedures

  • Cross-reactivity with other PDZ-containing proteins:

    • Challenge: SDCBP antibodies may cross-react with structurally similar proteins

    • Solution: Validate antibody specificity using SDCBP knockdown tissues/cells as negative controls; perform Western blot analysis to confirm single band at expected molecular weight

  • Tissue preservation issues:

    • Challenge: Formalin fixation can mask epitopes and reduce antibody binding

    • Solution: Optimize fixation time (24-48 hours); test multiple antigen retrieval methods (heat-induced vs. enzymatic)

  • Quantification difficulties:

    • Challenge: Subjective interpretation of staining intensity

    • Solution: Implement standardized scoring systems (negative, weak, moderate, strong) as used in published studies ; employ digital image analysis for objective quantification

How should researchers interpret contradictory SDCBP expression data between different detection methods?

When faced with contradictory SDCBP expression data between different detection methods, researchers should follow this systematic interpretation approach:

  • Consider method-specific limitations:

    • Immunohistochemistry provides spatial information but may be less quantitative

    • Western blotting offers quantitative data but loses spatial context

    • qPCR measures mRNA which may not correlate perfectly with protein levels

  • Evaluate technical factors:

    • Antibody clone specificity: Different antibody clones may recognize distinct epitopes

    • Protocol optimization: Suboptimal conditions in one method may cause false negatives

    • Sample preparation: Protein extraction methods may affect SDCBP detection

  • Biological considerations:

    • Post-translational modifications: These may affect antibody recognition in certain assays

    • Subcellular localization: SDCBP predominantly localizes in the cytoplasm in glioma cells , which may affect detection by certain methods

    • Protein-protein interactions: Binding partners may mask epitopes in certain contexts

  • Resolution strategy:

    • Employ additional orthogonal methods (e.g., mass spectrometry)

    • Use multiple antibody clones targeting different epitopes

    • Perform functional validation through knockdown/overexpression studies similar to the SDC1 manipulation experiments in U251 cells

    • Consider examining SDCBP at both mRNA and protein levels simultaneously

What are the key considerations for analyzing SDCBP expression correlation with clinical outcomes in glioma?

When analyzing SDCBP expression correlation with clinical outcomes in glioma, researchers should consider:

How does SDCBP compare with other biomarkers in predicting glioma progression?

Current research on SDCBP as a biomarker for glioma progression reveals several important comparisons with other established markers:

  • Expression pattern advantage:

    • SDCBP shows a clear positive correlation with malignant grade of glioma (rs=0.661, P<0.001), with expression increasing from non-tumorous brain tissues to low-grade and high-grade gliomas

    • This gradual increase makes SDCBP potentially more valuable than binary markers that may only distinguish tumor from non-tumor

  • Functional relationship to invasion:

    • SDCBP acts as an important mediator of invasion in glioma , directly connecting the biomarker to a key biological process in cancer progression

    • This functional link provides mechanistic insight beyond purely correlative biomarkers

  • Association with established pathways:

    • SDCBP interacts with the Rac1-STAT3 signaling axis and affects MMP2/MMP9 expression , connecting it to well-established cancer progression pathways

    • Researchers should compare SDCBP expression with these downstream effectors to establish predictive patterns

  • Complementary biomarker potential:

    • The positive correlation between SDCBP and SDC1 expression in glioma tissues (rs=0.628, P=0.001) suggests these markers could be used in combination

    • Researchers should investigate multi-marker panels including SDCBP to improve predictive accuracy

  • Methodological considerations:

    • When comparing SDCBP with other biomarkers, standardize detection methods across markers

    • Employ multivariate analysis to determine independent prognostic value

    • Use receiver operating characteristic (ROC) curve analysis to compare sensitivity and specificity

What are the most promising approaches for targeting the SDCBP pathway in experimental glioma models?

Several promising approaches for targeting the SDCBP pathway in experimental glioma models warrant further investigation:

  • RNA interference strategies:

    • siRNA-mediated knockdown of SDCBP, similar to the approach used for SDC1 in U251 cells

    • Design and validation of shRNA constructs for stable SDCBP suppression in long-term experiments

    • CRISPR-Cas9 genome editing to create SDCBP knockout glioma cell lines

  • Disruption of protein-protein interactions:

    • Development of small molecule inhibitors targeting SDCBP's PDZ domains

    • Design of peptide mimetics that compete with natural binding partners

    • Screening of compound libraries for molecules that disrupt SDCBP-SDC1 interaction

  • Antibody-based approaches:

    • Function-blocking SDCBP monoclonal antibodies that can penetrate cells

    • Consideration of BBB penetration strategies, as therapeutic antibodies typically show limited brain concentration (0.1-0.4% of serum levels)

    • Transferrin conjugation techniques that don't alter binding properties to target proteins

  • Pathway modulation strategies:

    • Targeting downstream effectors like Rac1 or STAT3 phosphorylation

    • Inhibition of MMP2/MMP9 expression or activity to block invasion capabilities

    • Combined inhibition of multiple nodes in the SDC1-SDCBP-Rac1-STAT3 signaling axis

  • Preclinical model considerations:

    • Validation in both in vitro 3D invasion models and orthotopic xenograft models

    • Assessment of effects on both tumor growth and invasive capacity

    • Combination with standard-of-care treatments to identify synergistic approaches

What new methodologies are emerging for studying SDCBP function in the tumor microenvironment?

Emerging methodologies for studying SDCBP function in the tumor microenvironment include:

  • Advanced imaging techniques:

    • Super-resolution microscopy to visualize SDCBP localization at the nanoscale level

    • Intravital microscopy using fluorescently tagged SDCBP antibodies to monitor protein dynamics in live animal models

    • Correlative light and electron microscopy (CLEM) to connect SDCBP localization with ultrastructural features

  • 3D culture systems:

    • Patient-derived organoids incorporating glioma cells and microenvironmental components

    • Bioprinted 3D models with controlled spatial organization of tumor and stromal cells

    • Microfluidic devices that allow real-time monitoring of SDCBP-expressing cells interacting with the microenvironment

  • Single-cell analysis approaches:

    • Single-cell RNA sequencing to identify SDCBP expression heterogeneity within tumors

    • Mass cytometry (CyTOF) with SDCBP antibodies to quantify protein levels at single-cell resolution

    • Spatial transcriptomics to map SDCBP expression patterns in relation to microenvironmental features

  • Proteomics advancements:

    • Proximity-dependent biotin identification (BioID) to map the SDCBP interactome in glioma cells

    • Phosphoproteomics to identify signaling networks affected by SDCBP expression

    • SILAC-based quantitative proteomics to measure changes in the secretome of SDCBP-manipulated glioma cells

  • In situ analysis techniques:

    • Multiplexed immunofluorescence to simultaneously visualize SDCBP with multiple markers

    • Digital spatial profiling to quantify SDCBP expression in precise regions of the tumor microenvironment

    • CODEX (CO-Detection by indEXing) for highly multiplexed protein mapping in tissue sections

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.