CDG1 Antibody

Shipped with Ice Packs
In Stock

Description

CDCP1 (CUB Domain-Containing Protein 1) Antibodies in Cancer Therapy

CDCP1 is a transmembrane glycoprotein overexpressed in multiple cancers, making it a rational target for antibody-drug conjugates (ADCs) and imaging agents.

Key Research Findings:

  • Diagnostic and Therapeutic Potential:

    • CDCP1 antibodies (e.g., ch10D7) demonstrate high specificity for cancer cells, enabling targeted delivery of cytotoxic agents like monomethyl auristatin E (MMAE) or radioisotopes (e.g., ⁸⁹Zr) .

    • Preclinical studies show ch10D7-MMAE conjugates induce robust anti-tumor effects in pancreatic, colorectal, and ovarian cancer xenografts .

Mechanisms of Action:

MechanismDescription
Payload DeliveryMMAE-linked antibodies trigger apoptosis in CDCP1-positive cancer cells .
Imaging⁸⁹Zr-labelled ch10D7 enables PET imaging for tumor detection in mice .
InternalizationCDCP1-antibody complexes undergo rapid internalization, enhancing drug uptake .

Clinical Relevance:

  • CDCP1 is highly expressed in pancreatic ductal adenocarcinoma, ovarian clear cell carcinoma, and prostate cancer, with limited expression in normal tissues .

  • Antibodies like ch10D7 and 4A06 show promise for personalized cancer treatment .

ALG1-CDG and Antibody Deficiency

ALG1-CDG (Congenital Disorder of Glycosylation type Ik) is a rare genetic disorder caused by mutations in the ALG1 gene, leading to impaired glycosylation and reduced immunoglobulin production.

Key Clinical Features:

  • Immunological Deficits:

    • Affected individuals exhibit hypogammaglobulinemia, particularly low IgG levels, increasing susceptibility to infections .

    • Impaired antibody function exacerbates clotting disorders and recurrent infections .

Pathogenesis:

  • ALG1 mutations disrupt mannose transfer during oligosaccharide synthesis, compromising glycoprotein function .

CDG1 Kinase in Plant Biology

In Arabidopsis thaliana, CDG1 (CONSTITUTIVE DIFFERENTIAL GROWTH1) is a receptor-like cytoplasmic kinase regulating brassinosteroid signaling and immune responses.

Functional Insights:

  • Brassinosteroid Signaling:

    • CDG1 interacts with BSU1 phosphatase to dephosphorylate BZR1 transcription factors, promoting growth .

  • Immune Regulation:

    • CDG1 negatively regulates pattern-triggered immunity by promoting degradation of immune receptors FLS2 and CERK1 .

Antibody Tools in Research:

  • Anti-CDCP1 antibodies (e.g., ch10D7) are distinct from plant CDG1 kinase studies, highlighting the importance of context in interpreting "CDG1 Antibody" .

Comparative Analysis of CDG1-Related Targets

TargetFunctionAntibody ApplicationKey References
CDCP1Cancer cell surface receptorDrug delivery, imaging
ALG1Glycosylation enzymeN/A (antibody deficiency)
CDG1Plant kinaseKinase activity studies

Future Directions

  • CDCP1-Targeted Therapies: Optimizing antibody half-life and payload efficiency could enhance clinical outcomes .

  • ALG1-CDG Management: IgG replacement therapy remains critical for addressing antibody deficiencies .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
CDG1 antibody; At3g26940 antibody; MOJ10.2Serine/threonine-protein kinase CDG1 antibody; EC 2.7.11.1 antibody; Protein CONSTITUTIVE DIFFERENTIAL GROWTH 1 antibody
Target Names
CDG1
Uniprot No.

Target Background

Function

This antibody targets CDG1, a serine/threonine-protein kinase crucial for the positive regulation of brassinosteroid (BR) signaling and subsequent plant growth. CDG1 acts as a mediator in BR signal transduction, relaying signals from the BRI1 receptor kinase to the BSU1 phosphatase. Specifically, following phosphorylation at Ser-234 by BRI1, CDG1 phosphorylates BSU1 at Ser-764 within its phosphatase domain. This phosphorylation enhances BSU1's ability to dephosphorylate ASK7/BIN2 (a negative regulator of BR signaling) at Tyr-200, thereby inactivating it. Full kinase activity of CDG1 is essential for its biological function.

Gene References Into Functions

Key functional aspects of CDG1 are supported by the following research:

  1. CDG1 mediates signal transduction from BRI1 to BSU1. PMID: 21855796
  2. Overexpression of the CDG1 gene may interfere with BR signal transduction; however, CDG1 is not essential for BR signaling in wild-type plants. PMID: 15466232
Database Links

KEGG: ath:AT3G26940

STRING: 3702.AT3G26940.1

UniGene: At.37130

Protein Families
Protein kinase superfamily, Ser/Thr protein kinase family
Subcellular Location
Cell membrane; Lipid-anchor.
Tissue Specificity
Expressed at high levels in the stamen and pollen grains. Expressed at a very low level in vegetative tissues.

Q&A

What is the relationship between CDG1 and antibodies used in glycosylation research?

CDG1 represents Type I Congenital Disorders of Glycosylation, genetic conditions affecting the assembly or transfer of lipid-linked oligosaccharide precursors. Antibodies targeting proteins involved in these pathways, such as CDGIE (a synonym of the DPM1 gene), are essential research tools for detecting and measuring relevant antigens in biological samples. The DPM1 gene encodes dolichyl-phosphate mannosyltransferase subunit 1, which functions in glycosylation and other metabolic processes . These antibodies enable fundamental research into glycosylation mechanisms and serve as diagnostic tools for identifying potential CDG cases through detection of abnormally glycosylated proteins.

What are the primary applications of anti-CDGIE/DPM1 antibodies in CDG research?

Anti-CDGIE antibodies have several critical applications in CDG research including Western Blotting, ELISA, and Immunohistochemistry . Western Blotting allows researchers to detect and quantify CDGIE/DPM1 protein levels across different experimental conditions, enabling correlation between protein expression and glycosylation defects. ELISA applications provide quantitative measurement of the antigen in various biological samples. Immunohistochemistry applications are valuable for determining tissue localization patterns, as CDGIE/DPM1 is reported to be localized primarily in the endoplasmic reticulum and is widely expressed across multiple tissue types . Together, these applications enable comprehensive investigation of glycosylation pathway components in both research and diagnostic contexts.

How do researchers select the appropriate CDGIE/DPM1 antibody for their experimental needs?

Selection of appropriate antibodies requires consideration of multiple experimental parameters. First, researchers should evaluate antibody reactivity profiles—available anti-DPM1 antibodies demonstrate specificity for human, mouse, and rat antigens in various combinations . Second, the intended application dictates selection; for example, some antibodies are optimized specifically for Western Blot and ELISA, while others may be validated for additional applications like immunohistochemistry. Third, antibody format matters—researchers can choose between unconjugated antibodies and those with specific conjugates depending on their detection system. Finally, researchers should evaluate antibody origin (e.g., goat anti-DPM1 antibodies) to ensure compatibility with their secondary detection systems and to avoid cross-reactivity in multi-labeling experiments .

What biomarkers do CDG1 antibodies detect for diagnostic purposes?

CDG1 antibodies target proteins critical to detecting several diagnostic biomarkers. The most established biomarker is carbohydrate-deficient transferrin (CDT), with at least one abnormal CDT test result being a key inclusion criterion for ALG1-CDG diagnosis . Antibodies enable detection of hypoglycosylation patterns in transferrin through techniques like Western blotting. Additionally, researchers have identified a novel protein-linked xeno-tetrasaccharide biomarker, NeuAc-Gal-GlcNAc₂, which was detected in all twenty-seven ALG1-CDG patients tested in a comprehensive study . This biomarker represents a significant advancement in diagnostic specificity. A third biomarker utilized in yeast complementation assays is carboxypeptidase Y (CPY) glycosylation, which provides a functional readout for assessing the pathogenicity of ALG1 mutations .

How are antibody-based assays used to differentiate between CDG subtypes?

Antibody-based assays are crucial for differentiating between the more than one hundred known CDG subtypes. For type I CDGs specifically, antibodies enable detection of characteristic glycosylation patterns that differ between subtypes. In ALG1-CDG, antibodies detecting the abnormal mobility of glycoproteins (due to incomplete glycan structures) help distinguish it from other CDG subtypes . Researchers employ transferrin Western blots to identify type I pattern abnormalities, followed by molecular genetic analysis to confirm specific gene defects. More advanced approaches combine antibody detection of biomarkers with mass spectrometry analysis of glycan structures, enabling precise differentiation between closely related CDG subtypes. The molecular analysis of ALG1-CDG has revealed 31 potential mutations across 39 affected individuals, with 26 of these mutations (84%) being previously unreported .

What are the technical considerations when using antibodies for screening potential CDG1 cases?

When screening for CDG1 disorders using antibodies, researchers must address several technical challenges. First, sample preparation is critical—consistent protein extraction methods must be employed to ensure reliable results across different tissues or patient samples. Second, researchers should include appropriate controls, particularly samples from confirmed CDG1 cases alongside normal controls, to establish valid reference points. Third, the selection of detection systems significantly impacts sensitivity; enhanced chemiluminescence systems may be required for detecting subtle glycosylation abnormalities. Fourth, quantification methods must be standardized, typically using densitometry with internal loading controls. Finally, researchers should consider validation through orthogonal methods—31 potential mutations in ALG1-CDG cases were identified using a combination of exome sequencing, targeted gene panels, and Sanger sequencing to ensure accuracy .

How can antibodies be used in conjunction with genetic analysis to study CDG1 pathophysiology?

The integrated use of antibodies with genetic analysis has revolutionized CDG1 research. Initially, antibody-based detection of abnormal glycosylation patterns can serve as a screening tool to identify potential CDG cases. Once identified, targeted genetic analysis (through Sanger sequencing, gene panels, or whole exome sequencing) can precisely define the underlying mutations . This dual approach is exemplified in ALG1-CDG research, where antibody-detected glycosylation abnormalities led to the discovery of 31 potential mutations across 39 patients from 32 families . After identifying mutations, researchers employ functional studies using antibodies to assess the impact of these mutations on protein expression, localization, and enzymatic function. This combined approach provides comprehensive insights into genotype-phenotype correlations, as demonstrated in the identification of mutations with lethal outcomes in the first two years of life .

What functional assays incorporate CDG1-related antibodies to assess pathogenicity of novel mutations?

Researchers have developed sophisticated functional assays incorporating antibodies to assess mutation pathogenicity in CDG1 disorders. A primary approach is the yeast complementation assay, where human ALG1 variants are expressed in alg1-deficient yeast strains and their functionality is assessed through monitoring growth restoration and CPY glycosylation using antibody detection . This method confirmed pathogenicity for all mutations tested in a study of 39 ALG1-CDG patients . Another critical approach employs antibodies in cell-based assays to detect altered glycosylation patterns in patient fibroblasts or transfected cell lines expressing mutant proteins. Immunofluorescence microscopy using antibodies against DPM1/CDGIE enables assessment of subcellular localization changes resulting from mutations. Additionally, antibodies facilitate co-immunoprecipitation experiments to evaluate disrupted protein-protein interactions within the glycosylation machinery, providing mechanistic insights into pathogenicity.

How are antibodies utilized in investigating the molecular mechanisms of different CDG1 subtypes?

Antibodies serve as critical tools for elucidating the molecular mechanisms underlying various CDG1 subtypes. In ALG1-CDG, antibodies enable precise characterization of the β1,4 mannosyltransferase function, which catalyzes the addition of the first mannose moiety in N-linked glycosylation . Through immunoprecipitation followed by activity assays, researchers can directly measure enzymatic function affected by specific mutations. Antibodies also enable protein expression profiling across different tissues and developmental stages, providing insights into the tissue-specific manifestations of CDG1 disorders. Additionally, researchers employ antibodies in multiplexed immunofluorescence to visualize colocalization patterns of glycosylation machinery components, revealing how specific mutations disrupt the spatial organization of the pathway. These approaches have contributed to understanding why certain mutations, such as homozygous p.Ser258Leu in ALG1-CDG, result in severe phenotypes with early mortality .

What are the critical controls required when using antibodies to study CDG1-related proteins?

Proper experimental controls are essential when studying CDG1-related proteins with antibodies. First, researchers must include positive controls—well-characterized cell lines or tissue samples known to express the target protein at detectable levels. Second, negative controls are equally important, including samples from knockout models or cell lines where the target protein is absent or significantly reduced. Third, isotype controls using non-specific antibodies of the same isotype help distinguish specific binding from background signals. Fourth, knockdown/knockout validation experiments, where antibody signal disappears or diminishes following genetic silencing of the target, confirm detection specificity. Finally, for functional studies like the yeast complementation assay used to validate ALG1 mutations, both wild-type complementation (positive control) and empty vector (negative control) conditions are essential benchmarks .

What protocols are recommended for optimizing Western blot detection of hypoglycosylated proteins in CDG1 samples?

ParameterRecommendationRationale
Sample preparationGentle lysis with glycosidase inhibitorsPreserves native glycosylation status
Protein loading25-50 μg total proteinEnsures detection of low-abundance glycoproteins
Gel percentage7-10% SDS-PAGEOptimal separation of glycoforms
Transfer conditionsWet transfer, 30V overnight at 4°CEfficient transfer of high molecular weight glycoproteins
Blocking solution5% BSA in TBS-TReduces background without binding to glycan epitopes
Primary antibody dilution1:1000-1:5000 (optimize per antibody)Balance between signal strength and specificity
Detection methodEnhanced chemiluminescenceProvides sensitivity needed for subtle glycoform differences
ControlsInclude known CDG samples and healthy controlsEssential for interpretation of abnormal patterns

This protocol has been optimized based on extensive experience with transferrin Western blots in CDG research and can be adapted for other glycoproteins relevant to CDG1 investigation .

How can researchers overcome technical challenges when using antibodies against highly conserved glycosylation pathway proteins?

Detecting highly conserved glycosylation pathway proteins presents several technical challenges. First, researchers should prioritize using antibodies raised against species-specific epitopes—commercial anti-DPM1 antibodies are available with validated reactivity against human, mouse, or rat antigens, allowing species-appropriate selection . Second, epitope masking due to protein-protein interactions in the glycosylation machinery can be addressed through optimized sample preparation including mild detergents and denaturing conditions where appropriate. Third, cross-reactivity with homologous proteins can be minimized by pre-absorbing antibodies against recombinant related proteins or through careful epitope selection during antibody development. Fourth, researchers should employ orthogonal validation techniques such as mass spectrometry to confirm antibody specificity. Finally, when working with highly conserved proteins like dolichyl-phosphate mannosyltransferase, researchers can use multiple antibodies targeting different epitopes to increase confidence in detection specificity .

How are antibodies being used to develop potential therapeutic approaches for CDG1 disorders?

Antibodies are facilitating multiple therapeutic research directions for CDG1 disorders. First, they enable high-throughput screening of small molecule libraries to identify compounds that may correct specific glycosylation defects, with antibody-based detection of glycosylation status serving as the readout. Second, therapeutic antibodies conjugated with enzyme replacement payloads are being explored as targeted delivery systems for correcting enzymatic deficiencies in specific tissues. Third, antibodies enable patient stratification for clinical trials by precisely characterizing the molecular defects present in individual patients. Fourth, antibody-based imaging allows researchers to track the distribution and efficacy of experimental therapies in animal models. Finally, biomarker antibodies are crucial for monitoring treatment responses in both preclinical models and potential clinical trials, paralleling the theranostic approach developed for other conditions like those targeting CDCP1 in cancer research .

What are the latest developments in antibody-based imaging techniques for studying CDG1 pathophysiology?

Recent advances in antibody-based imaging are transforming CDG1 research. Super-resolution microscopy using fluorophore-conjugated antibodies now allows visualization of glycosylation machinery components at nanometer resolution, revealing previously undetectable organizational defects in CDG1 patient cells. Multiplexed immunofluorescence techniques enable simultaneous visualization of multiple glycosylation pathway components, providing insights into their spatial relationships within the secretory pathway. Antibody-based proximity labeling methods like BioID and APEX2 are being applied to map the protein interaction networks disrupted in various CDG1 subtypes. In animal models, advances in antibody-based in vivo imaging parallel developments in other fields, such as the 89Zirconium-labelled antibody that enables PET imaging of target proteins . Additionally, antibody-based tissue clearing methods are enabling three-dimensional visualization of glycosylation defects across intact tissues, providing organ-level insights into CDG1 pathophysiology.

How do researchers correlate in vitro antibody-based findings with clinical phenotypes in CDG1 patients?

Correlating laboratory findings with clinical manifestations represents one of the most challenging aspects of CDG1 research. Researchers employ several methodologies to establish these correlations. First, they conduct comprehensive phenotyping of patient cohorts alongside molecular and antibody-based analyses, as exemplified in the study of 39 ALG1-CDG patients where detailed clinical information was collected alongside genetic and biochemical data . Second, researchers utilize patient-derived fibroblasts or induced pluripotent stem cells for in vitro studies, allowing direct link between patient-specific mutations and cellular phenotypes detected with antibodies. Third, severity scoring systems for clinical manifestations can be correlated with quantitative measures of protein glycosylation detected by antibodies. Fourth, longitudinal studies track changes in biomarkers detected by antibodies alongside clinical progression. Notably, researchers identified mutations with lethal outcomes in the first two years of life and correlated these with specific glycosylation patterns , demonstrating the value of integrated clinical-molecular approaches.

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.