OPTC Antibody

Shipped with Ice Packs
In Stock

Description

Expression in Hematological Malignancies

  • CLL and MCL: OPTC mRNA and protein are universally expressed in CLL patients (n = 90) and detected in 25% of mantle cell lymphoma (MCL) cases (2/8 patients) .

  • Specificity: Absent in healthy peripheral blood mononuclear cells (PBMCs; n = 20) and nine other hematological malignancies .

Subcellular Localization

  • In CLL cells, OPTC localizes to the nucleus and endoplasmic reticulum (ER), contrasting with its typical ECM presence in normal tissues like the eye .

Clinical Correlations

ObservationData
Expression in progressive vs. non-progressive CLLHigher mean OPTC expression in progressive cases (n = 16 vs. n = 14), but not statistically significant .
Diagnostic utilityOPTC serves as a potential biomarker for CLL due to tumor-specific expression .

Functional Insights

  • Anti-Angiogenic Role: In ocular tissues, OPTC inhibits α2β1 and α1β1 integrin-mediated endothelial cell adhesion to collagen, suppressing angiogenesis .

  • Cancer Biology: SLRPs like OPTC may antagonize tumor growth and metastasis, though their role in CLL remains under investigation .

Technical Considerations

  • Positive Controls: CLL patient samples or TM3 mouse cells expressing human OPTC .

  • Negative Controls: Healthy donor PBMCs or non-CLL hematological malignancy samples .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery time may vary depending on your location and shipping method. Please contact your local distributor for specific delivery timelines.
Synonyms
Oculoglycan antibody; OPT antibody; OPT_HUMAN antibody; OPTC antibody; Opticin antibody
Target Names
OPTC
Uniprot No.

Target Background

Function
Opticin plays a crucial role in inhibiting angiogenesis within the vitreous humor of the eye, effectively suppressing neovascularization. It binds to collagen fibrils and potentially contributes to collagen fiber organization by regulating other members of the small leucine-rich repeat proteoglycan superfamily.
Gene References Into Functions
  1. Opticin binds to collagen, competitively inhibiting endothelial cell interactions with collagen via alpha(1)beta(1) and alpha(2)beta(1) integrins. This inhibition prevents the strong adhesion necessary for proangiogenic signaling through these integrins. PMID: 22669977
  2. VEGF and hypoxia can decrease opticin protein levels in the human retinal pigment epithelium secretome. Additionally, opticin may serve as an enzymatic substrate for MMP-2. PMID: 22534113
  3. Research has investigated the localization of opticin in the human eye and conducted genetic screens. PMID: 12019215
  4. OPTC has been identified as a candidate gene for primary open-angle glaucoma. PMID: 17359525
  5. Studies suggest that OPTC and EPYC are unlikely to play a significant role in high myopia. PMID: 19844586

Show More

Hide All

Database Links

HGNC: 8158

OMIM: 605127

KEGG: hsa:26254

STRING: 9606.ENSP00000356191

UniGene: Hs.632468

Protein Families
Small leucine-rich proteoglycan (SLRP) family, SLRP class III subfamily
Subcellular Location
Secreted, extracellular space, extracellular matrix.
Tissue Specificity
Expressed in cartilage and synovial membranes (at protein level). Expressed in the retina, iris, ligament, skin and fetal liver (at protein level). Expressed in the retinal pigment epithelium (at protein level). Expressed in synovial fibroblasts and subch

Q&A

What is OPTC and why is it significant in antibody research?

OPTC (opticin) is a member of the small leucine-rich proteoglycan (SLRP) family that has gained attention in cancer research due to its differential expression patterns. In normal physiology, OPTC is a glycosylated protein running at 45-50 kDa on SDS-PAGE gels and is secreted into the extracellular matrix (ECM) of various tissues including the eye, brain, skin, ligament, and cartilage . Its significance in antibody research stems from its unique expression profile in certain hematological malignancies, particularly chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL), where it has been found to be overexpressed compared to healthy controls . This differential expression makes OPTC a potential biomarker and therapeutic target, driving research into developing and characterizing antibodies against this protein. The research significance is further underscored by findings that extracellular OPTC protein exhibits anti-angiogenic properties by binding to collagen and inhibiting integrin-mediated endothelial cell adhesion, as well as potentially modulating growth hormone function .

How do researchers validate the specificity of anti-OPTC antibodies?

Validating anti-OPTC antibody specificity requires a multi-step process to ensure reliable experimental outcomes. According to the literature, researchers typically employ Western blot analysis against recombinant OPTC proteins expressed in different systems. For example, researchers have tested C-terminal anti-OPTC polyclonal antibodies against recombinant bovine OPTC and human OPTC expressed in E. coli and TM3 mouse cells, with cells transfected with vector alone serving as negative controls . In these validation studies, bovine OPTC was recognized as a 45-50 kDa band, corresponding to the mature glycosylated protein, while human OPTC expressed in TM3 mouse cells appeared as a 48 kDa band . Additionally, OPTC expressed in bacterial systems (JM109) was detected as a 37-38 kDa band, representing the unglycosylated form . Comprehensive validation also involves confirming that both N-terminal and C-terminal antibodies recognize the same protein to rule out degradation or cleavage products. For instance, the 37 kDa band found in CLL patients was recognized by both C-terminal and N-terminal polyclonal antibodies, suggesting it was likely a full-length unglycosylated protein rather than a fragment .

What are the key differences between OPTC antibodies and other commonly studied antibodies in neurological disorders?

OPTC antibodies differ significantly from other commonly studied antibodies in neurological disorders, particularly MOG (myelin oligodendrocyte glycoprotein) and AQP4 (aquaporin-4) antibodies, in terms of target tissue, clinical associations, and detection methodologies. While OPTC is primarily associated with hematological malignancies like CLL and MCL , MOG and AQP4 antibodies are primarily implicated in neurological disorders such as MOGAD and neuromyelitis optica spectrum disorders (NMOSD) . Detection methodologies also differ significantly. For OPTC antibodies, Western blot analysis of cell lysates separated into cytosolic and membrane/organelle fractions is commonly employed, with the 37 kDa unglycosylated OPTC protein typically identified in the Triton X-resistant fraction containing membrane and organelle proteins . In contrast, MOG and AQP4 antibodies are typically detected using cell-based assays (CBAs), with MOG antibodies detected using either live or fixed cell-based assays, and flow cytometry being used to quantify binding with specific cutoff values (e.g., ≥100 considered high positive, 20-40 considered low positive for MOG antibodies) . Clinical interpretation also differs substantially: while low-titer MOG antibodies may be found in 1-2% of multiple sclerosis patients and can confuse diagnosis , OPTC expression appears to be more specifically associated with certain leukemic conditions and absent in healthy controls .

How do researchers quantify OPTC expression in experimental samples?

Researchers quantify OPTC expression through multiple complementary techniques to ensure accurate measurements across different experimental contexts. The primary method documented in the literature is quantitative PCR (qPCR), where OPTC expression is measured relative to housekeeping genes such as GAPDH . This approach allows for statistical comparison of expression levels between different sample groups, such as CLL patients versus healthy controls, or progressive versus non-progressive CLL patients . Western blot analysis serves as a complementary protein-level quantification method, with researchers typically employing a two-step cell lysis method that separates cytosolic proteins from membrane and organelle proteins . This fractionation approach has revealed that the 37 kDa unglycosylated OPTC protein in CLL cells is predominantly located in the Triton X-resistant fraction containing membrane and organelle proteins, rather than in the cytosolic fraction . For more detailed subcellular localization studies, additional techniques such as immunofluorescence microscopy would be appropriate, though specific protocols for OPTC were not detailed in the provided search results. When comparing expression across patient groups, researchers have found that the relative expression of OPTC is significantly higher in CLL patients compared to healthy controls (p<0.0001), regardless of whether the patients had progressive or non-progressive disease, though the mean relative expression appeared somewhat higher in progressive CLL patients .

What are the challenges in developing monoclonal antibodies against different epitopes of OPTC?

Developing monoclonal antibodies against different OPTC epitopes presents several technical challenges due to the protein's structural characteristics and expression patterns. First, the dual existence of OPTC as both glycosylated (45-50 kDa) and unglycosylated (37 kDa) forms means that epitope accessibility may vary significantly between these variants . Glycosylation can mask potential antigenic determinants, making it difficult to develop antibodies that consistently recognize both forms with equal affinity. Second, the small leucine-rich proteoglycan (SLRP) family, to which OPTC belongs, contains several structurally homologous proteins, creating potential cross-reactivity issues . Researchers must carefully select unique epitopes and extensively validate antibody specificity against other SLRP family members to ensure target specificity. Third, the differential subcellular localization of OPTC forms—with unglycosylated forms found preferentially in the nucleus and endoplasmic reticulum of CLL cells—means that antibodies must be validated for their ability to recognize OPTC in different cellular compartments and under various fixation and permeabilization conditions . This becomes particularly important for immunohistochemistry or immunofluorescence applications where fixation methods might alter epitope conformation. Additionally, the relatively low expression of OPTC in normal tissues compared to its overexpression in certain malignancies means that antibodies must have excellent sensitivity to detect physiological expression levels while maintaining specificity at the higher concentrations found in disease states .

How can researchers distinguish between false positive and true positive OPTC antibody signals in complex biological samples?

Distinguishing between false positive and true positive OPTC antibody signals in complex biological samples requires a multi-faceted validation approach to ensure experimental reliability. First, researchers should implement rigorous positive and negative controls in every experimental setup. As demonstrated in the literature, cells transfected with vector alone serve as essential negative controls, while cells expressing recombinant OPTC (such as TM3 mouse cells transfected with human OPTC) function as positive controls . Second, researchers should validate findings using multiple antibodies targeting different epitopes of OPTC. The research shows that both N-terminal and C-terminal antibodies should recognize the same protein band to confirm specificity; divergent recognition patterns might indicate degradation products or cross-reactivity . Third, competitive binding assays, where pre-incubation with purified recombinant OPTC protein blocks specific antibody binding, can help differentiate specific from non-specific signals. Fourth, comparative analysis across multiple detection methods strengthens result validity—for instance, combining Western blot findings with qPCR data showing OPTC overexpression at the transcriptional level in the same samples . Finally, researchers should establish clear threshold criteria based on signal-to-noise ratios specific to their experimental system. In MOG antibody research, for example, specific cutoff values (≥100 for high positive, 20-40 for low positive) help distinguish genuine from borderline results . This approach could be adapted for OPTC antibody detection to reduce ambiguity in signal interpretation.

How should researchers approach epitope mapping for OPTC antibodies?

Epitope mapping for OPTC antibodies requires a systematic approach that accounts for the protein's structural features and post-translational modifications. Researchers should begin with in silico analysis of the OPTC sequence to identify potentially immunogenic regions, paying particular attention to areas that display high surface accessibility and low sequence homology with other SLRP family members to minimize cross-reactivity . Following computational prediction, researchers should employ peptide arrays using overlapping synthetic peptides spanning the entire OPTC sequence to experimentally verify antibody binding sites. This approach allows for precise identification of linear epitopes. For conformational epitopes, hydrogen-deuterium exchange mass spectrometry (HDX-MS) or X-ray crystallography of antibody-antigen complexes would provide structural insights into binding interfaces. Researchers must specifically compare epitope accessibility between the glycosylated 45-50 kDa and unglycosylated 37 kDa forms of OPTC to understand how post-translational modifications affect antibody recognition . Additionally, mutagenesis studies introducing systematic alanine substitutions at predicted epitope residues can confirm the functional importance of specific amino acids for antibody binding. Competition assays between different antibody clones can further elucidate whether antibodies target overlapping or distinct epitopes, informing the development of antibody panels that collectively provide comprehensive coverage of the protein. Finally, cross-species reactivity testing is essential to determine whether identified epitopes are conserved across species, facilitating translational research from animal models to human applications.

What are the optimal sample preparation techniques for detecting OPTC in different tissue types?

Optimal sample preparation techniques for detecting OPTC vary significantly depending on tissue type, target application, and whether researchers aim to detect the glycosylated or unglycosylated form of the protein. For hematological samples, such as those from CLL or MCL patients, a two-step cell lysis method has proven effective in separating cytosolic proteins from membrane and organelle-associated proteins . This approach is crucial since the 37 kDa unglycosylated OPTC variant in CLL cells is predominantly found in the Triton X-resistant fraction containing membrane and organelle proteins, rather than in the cytosolic fraction . For solid tissues where OPTC is normally expressed (eye, brain, skin, ligament, and cartilage), researchers should consider implementing tissue-specific extraction buffers that effectively solubilize extracellular matrix components where the glycosylated 45-50 kDa OPTC typically resides . When preparing samples for immunohistochemistry or immunofluorescence, fixation protocols must be carefully optimized, as overfixation may mask epitopes while underfixation can compromise tissue morphology. For formalin-fixed paraffin-embedded (FFPE) tissues, antigen retrieval methods should be systematically compared to determine which approach best exposes OPTC epitopes without causing tissue degradation. Additionally, researchers should consider subcellular fractionation techniques that separately isolate nuclear, endoplasmic reticulum, and cell membrane components, since unglycosylated OPTC in CLL cells has been found to localize preferentially to the nucleus and endoplasmic reticulum . For proteomic applications, researchers must account for the different molecular weights of glycosylated versus unglycosylated OPTC forms when selecting gel separation parameters or mass spectrometry protocols .

How can contradictory OPTC antibody results between different experimental platforms be reconciled?

Reconciling contradictory OPTC antibody results between different experimental platforms requires systematic investigation of multiple variables that could contribute to discrepancies. First, researchers should examine antibody characteristics, including clone specificity, isotype, and epitope targets. Different antibodies may recognize distinct epitopes that are differentially accessible in various experimental conditions—antibodies targeting regions affected by glycosylation might detect the 45-50 kDa glycosylated form but miss the 37 kDa unglycosylated variant found in CLL cells . Second, sample preparation methods significantly impact results; the two-step cell lysis method revealing OPTC in the Triton X-resistant fraction but not in the cytosolic fraction of CLL cells demonstrates how extraction protocols influence detection outcomes . Third, platform-specific technical factors must be considered—Western blot denaturing conditions may expose epitopes hidden in native-condition ELISAs, while flow cytometry requires cell surface accessibility that may not reflect total cellular expression detected in lysate-based assays. Fourth, quantification methods and threshold definitions vary across platforms; standardized positive controls and calibration curves should be implemented across techniques to enable meaningful comparison of results. Fifth, cell or tissue types may exhibit different OPTC expression patterns and post-translational modifications; contradictory results might actually reflect true biological variability rather than technical artifacts . To systematically address these variables, researchers should implement cross-validation studies using multiple detection methods on identical samples, conduct spike-in recovery experiments to assess matrix effects, perform epitope mapping to understand which antibody recognizes which region of OPTC under different conditions, and establish standardized reporting frameworks that explicitly detail all methodological parameters to facilitate accurate comparison across laboratories and experimental platforms.

How can OPTC antibodies be utilized in monitoring disease progression in hematological malignancies?

OPTC antibodies present a promising tool for monitoring disease progression in hematological malignancies, particularly in CLL and MCL, where OPTC has shown specific overexpression patterns. The research indicates that OPTC expression may correlate with disease progression, as the mean relative expression appeared somewhat higher in progressive (n=16) than non-progressive (n=14) CLL patients, though this difference did not reach statistical significance in the limited sample size studied . To develop effective monitoring protocols, researchers should first establish standardized quantitative assays that reliably detect the 37 kDa unglycosylated OPTC variant characteristic of malignant cells . These assays could include quantitative flow cytometry to assess the percentage of OPTC-positive cells in peripheral blood or bone marrow samples, as well as quantitative immunohistochemistry to evaluate OPTC expression levels in tissue biopsies. For longitudinal monitoring, serial sampling protocols should be implemented, with samples collected at diagnosis, during treatment, and at regular follow-up intervals to track changes in OPTC expression over time. This approach would help determine whether changes in OPTC levels precede clinical disease progression, potentially serving as an early warning biomarker. Additionally, researchers should explore combining OPTC antibody-based detection with established prognostic markers to develop integrated risk assessment models that could improve prediction accuracy. Multiparametric analysis correlating OPTC expression levels with clinical outcomes across larger patient cohorts would help establish clinically relevant thresholds that distinguish stable from progressive disease. Finally, exploring whether qualitative changes in OPTC, such as altered subcellular localization patterns, correlate with disease evolution could provide additional prognostic information beyond simple expression level quantification .

What are the potential applications of OPTC antibodies in immunohistochemical analysis of tumor tissues?

OPTC antibodies offer several valuable applications in immunohistochemical analysis of tumor tissues, with potential for both diagnostic and research purposes. First, OPTC immunohistochemistry (IHC) could serve as a differential diagnostic tool to distinguish CLL and certain MCL cases from other hematological malignancies, given the specific expression pattern observed in these diseases but not in nine other types of hematological malignancies or healthy controls . This specificity could be particularly valuable in challenging cases where conventional markers yield ambiguous results. Second, OPTC IHC could potentially help identify tissue infiltration by CLL or MCL cells in extranodal sites, facilitating staging and treatment planning. Third, spatial analysis of OPTC expression patterns within tumor microenvironments could reveal heterogeneity in expression across different regions of the tumor, potentially correlating with local proliferative activity or other biological behaviors. Fourth, multiplex immunohistochemistry combining OPTC antibodies with other markers could help characterize the phenotypic profile of OPTC-expressing cells and their relationships with surrounding stromal and immune cells. For optimal implementation, researchers should develop standardized IHC protocols addressing epitope retrieval methods, antibody concentration, incubation conditions, and detection systems specific to formalin-fixed paraffin-embedded tissues. Scoring systems should be established to quantify OPTC expression levels, incorporating both intensity and distribution parameters. Additionally, artificial intelligence-based image analysis algorithms could be developed to provide objective and reproducible quantification of OPTC expression patterns across different tissue samples .

How might researchers design functional assays to study the biological significance of OPTC in disease pathogenesis?

Designing functional assays to study OPTC's biological significance in disease pathogenesis requires a multifaceted approach targeting different aspects of the protein's potential functions. First, researchers should develop gene knockdown and overexpression systems in relevant cell lines (such as CLL-derived cell lines) to manipulate OPTC levels and observe the resulting phenotypic changes . CRISPR-Cas9 gene editing could create stable OPTC knockout cell lines, while lentiviral vectors could establish overexpression models. Second, researchers should investigate the impact of OPTC on cellular processes relevant to cancer progression, including proliferation assays (MTT/XTT, BrdU incorporation), apoptosis assays (Annexin V/PI staining, caspase activation), and migration/invasion assays (transwell, wound healing). These functional readouts would help determine whether OPTC actively contributes to malignant behavior or represents a passive biomarker . Third, researchers should explore potential signaling pathways affected by OPTC, drawing from known functions of other SLRP family members. Phosphorylation arrays and protein-protein interaction studies using co-immunoprecipitation followed by mass spectrometry could identify OPTC binding partners and downstream effectors. Fourth, researchers should investigate the differential roles of glycosylated versus unglycosylated OPTC forms, potentially using site-directed mutagenesis to generate glycosylation-deficient variants and compare their functional impacts . Additionally, subcellular localization studies using fluorescently tagged OPTC variants could elucidate whether nuclear localization of the unglycosylated form in CLL cells has functional significance, potentially through chromatin immunoprecipitation sequencing (ChIP-seq) to identify any DNA binding activity . Finally, in vivo models employing xenografts of OPTC-manipulated cell lines in immunodeficient mice could assess the protein's impact on tumor formation, growth kinetics, and metastatic potential in a more physiologically relevant context.

What considerations are important when developing OPTC antibodies for potential therapeutic applications?

Developing OPTC antibodies for therapeutic applications requires addressing several critical considerations to ensure efficacy, safety, and target specificity. First, epitope selection must be strategically approached to target regions of OPTC that are consistently expressed and accessible in malignant cells but minimally exposed in normal tissues where OPTC is physiologically expressed (eye, brain, skin, ligament, and cartilage) . This differential targeting could potentially exploit the unglycosylated 37 kDa OPTC variant found specifically in CLL cells versus the 45-50 kDa glycosylated form in normal tissues . Second, antibody format selection must be carefully considered—full IgG antibodies offer longer half-life and effector functions like antibody-dependent cellular cytotoxicity (ADCC), while smaller formats such as Fab fragments or single-chain variable fragments (scFvs) may offer superior tissue penetration but shorter circulation time. Third, researchers must characterize potential on-target/off-tumor effects by comprehensively mapping OPTC expression across normal human tissues using sensitive detection methods, since physiological expression in critical organs could lead to adverse effects. Fourth, antibody engineering approaches should be explored to enhance therapeutic efficacy, including optimization of affinity (though not necessarily maximizing it, as ultra-high affinity can impair tissue penetration), Fc engineering to enhance or suppress specific effector functions as needed, and potential development of antibody-drug conjugates (ADCs) if internalization of OPTC occurs upon antibody binding. Fifth, researchers should evaluate combination strategies with standard-of-care treatments for CLL and MCL to assess potential synergistic effects. Finally, robust manufacturing processes must be developed that ensure consistent glycosylation and other post-translational modifications of the therapeutic antibody itself, as these characteristics significantly impact pharmacokinetics, immunogenicity, and biological activity of the final therapeutic product.

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 2024 Thebiotek. All Rights Reserved.