CD46 Antibody

CD46, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction to CD46 Antibody

CD46 (Cluster of Differentiation 46), also termed Membrane Cofactor Protein, is a ubiquitously expressed complement inhibitor that protects host cells from autologous complement attack by binding and inactivating C3b and C4b . CD46 antibodies are monoclonal or polyclonal reagents designed to target extracellular epitopes of CD46 for diagnostic or therapeutic purposes. Their development leverages CD46's overexpression in malignancies, including prostate cancer, multiple myeloma, and medulloblastoma .

Mechanisms of Action

CD46 antibodies exhibit dual functionality:

  • Complement Regulation: By binding CD46, antibodies modulate its cofactor activity for C3b/C4b inactivation, altering complement-mediated cell protection .

  • Pathogen and Therapeutic Internalization: CD46 serves as a receptor for measles virus, adenoviruses, and bacterial pathogens. Antibodies like YS5 exploit CD46's internalization properties to deliver cytotoxic payloads (e.g., MMAE/MMAF) into cancer cells .

Key Mechanistic Findings:

  • Anti-CD46 ADCs (e.g., FOR46) induce apoptosis by disrupting microtubule networks post-internalization .

  • CD46 engagement regulates T-cell differentiation, promoting IL-10-producing Tr1 cells, which may suppress inflammation .

Oncology

CD46 antibodies are central to ADC development:

ADC NameTarget CancerPayloadClinical PhaseKey FindingsSource
FOR46 (FG-3246)mCRPC, MyelomaMMAEPhase 1a/1b44.4% tumor regression (8/18 pts); 22.2% PR
CD46-ADCMultiple Myeloma (MM)MMAFPreclinicalEC₅₀ in picomolar range; eradicated MM xenografts
  • Prostate Cancer: In metastatic castration-resistant prostate cancer (mCRPC), CD46 is homogenously expressed in adenocarcinoma and neuroendocrine subtypes. FOR46, an anti-CD46 ADC, showed sustained tumor regression in 44.4% of patients in a Phase 1a trial .

  • Multiple Myeloma: CD46 antigen density on myeloma cells (454,668–470,991/cell) exceeds that of CD38, a standard MM marker. CD46-ADC eliminated orthotopic MM xenografts at 4 mg/kg dosing .

Inflammatory Diseases

CD46 deficiency or dysfunction correlates with impaired IL-10 production in multiple sclerosis, suggesting therapeutic potential in autoimmune disorders .

CD46 Expression Profiles

Cell TypeCD46 Antigen Density (Mean)NotesSource
Myeloma Cells454,668–470,991100% of patient samples (n=25)
Normal Plasma Cells22,475Low expression in non-malignant
Granulocytes (Normal)40,523–58,320Highest among normal hematopoietic

Preclinical Efficacy

  • Myeloma: CD46-ADC reduced tumor burden in NSG mice with MM1.S-Luc xenografts, achieving complete remission at 4 mg/kg .

  • Toxicity: Non-human primate studies showed acceptable safety profiles for CD46-targeted ADCs, with transient neutropenia as the primary adverse event .

Antibody Variants and Clones

CloneIsotypeApplicationsKey FeaturesSource
YS5IgG1ADC development (FOR46)High affinity for tumor CD46; internalizes via macropinocytosis
MEM-258IgG1Flow cytometry, pathogen studiesBinds adenovirus interaction site
TRA-2-10IgG2aT-cell regulation studiesRecognizes complement-binding domain
8E2IgG2aComplement inhibition assaysPE-conjugated; detects C3b/C4b binding

Future Directions

  • Combination Therapies: Pairing CD46 ADCs with androgen receptor inhibitors (e.g., enzalutamide) to enhance efficacy in mCRPC .

  • Alpha-Particle Therapy: ²¹²Pb-labeled YS5 demonstrated potent cytotoxicity in prostate cancer models, expanding CD46's therapeutic scope .

Product Specs

Introduction
Membrane cofactor protein isoform 14 (CD46) is a type I membrane protein that acts as a cofactor for the inactivation of complement components C3b and C4b by serum factor I. This function is located in the complement control protein repeats (CCPs), with CCP1-4 being crucial for regulation. CD46 is also involved in the fusion of sperm and egg during fertilization. CD46 is commonly found in thymocytes, T cells, B cells, monocytes, granulocytes, NK cells, platelets, endothelial cells, epithelial cells, fibroblasts, placenta, and sperm.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
1 mg/ml CD46 antibody in PBS, pH 7.4, with 10% glycerol and 0.02% sodium azide.
Storage Procedures
Store at 4°C for up to 1 month. For long-term storage, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
Stable for 12 months at -20°C or 1 month at 4°C.
Applications
CD46 antibody has been validated for ELISA and Western blot analysis to ensure specificity and reactivity. However, optimal working dilutions should be determined by titration for each application. Recommended starting dilution is 1:1000.
Synonyms
AHUS2, MCP, MIC10, TLX, TRA2.10, Membrane cofactor protein, Trophoblast leukocyte common antigen, CD46.
Purification Method
CD46 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT2G9AT.
Immunogen
Anti-human CD46 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human CD46 protein 35-313 amino acids purified from E. coli.
Ig Subclass
Mouse IgG2a heavy chain and k light chain.

Q&A

What is CD46 and what are its primary functions in cellular biology?

CD46, also known as Membrane Cofactor Protein (MCP) or MIC10, is a 45-70 kDa complement regulatory protein that serves as a protective mechanism for host cells. It functions by binding to C3b and C4b, allowing factor I (a serine protease) to degrade these components, thereby inhibiting complement activation on host tissues and preventing autologous attack. Additionally, CD46 serves as a receptor for measles virus. The protein contains four complement control protein domains (short consensus repeats or SCRs) and is expressed on virtually all nucleated cells, with the exception of erythrocytes .

What is the optimal experimental methodology for detecting CD46 across different applications?

Detection methodologies for CD46 vary by application:

Western Blot (WB):

  • Recommended dilution: 1:2000-1:14000

  • Sample preparation: Non-reduced sample treatment generally works best

  • Expected molecular weight: 50-56 kDa (though can range from 45-70 kDa)

  • Positive controls: A549, HeLa, Jurkat, and MOLT-4 cells

Immunohistochemistry (IHC):

  • Recommended dilution: 1:500-1:2000

  • Antigen retrieval: TE buffer (pH 9.0) or alternatively citrate buffer (pH 6.0)

  • Positive controls: Human ovary tumor, breast cancer, colon cancer, or cervical cancer tissues

Immunofluorescence/Immunocytochemistry (IF/ICC):

  • Recommended dilution: 1:50-1:500

  • Positive controls: HeLa cells

Immunoprecipitation (IP):

  • Recommended amount: 0.5-4.0 μg antibody for 1.0-3.0 mg total protein lysate

  • Positive controls: A549 cells

For all applications, it is essential to titrate the antibody in each specific experimental system to achieve optimal results .

How does CD46 protein heterogeneity impact antibody selection and experimental design?

CD46 exhibits significant heterogeneity that impacts experimental approaches:

  • Isoform variation: Four predominant isoforms arise from alternative splicing of a single CD46 gene, potentially affecting epitope availability .

  • Molecular weight variation: While the calculated molecular weight is approximately 43 kDa (384 amino acids), observed weights typically range from 50-56 kDa and can extend to 45-70 kDa due to post-translational modifications .

  • Epitope-specific antibodies: Some antibodies target specific domains, such as mAb M177 which recognizes the SCR2 domain .

When designing experiments, researchers should:

  • Select antibodies validated for their specific application

  • Consider using antibodies against conserved regions if detecting all isoforms is desired

  • Include appropriate controls to account for heterogeneity

  • Be aware that different epitope-specific antibodies may yield varying results depending on the accessibility of the target region in different experimental conditions

How does CD46 expression differ between normal and malignant cells, and what are the implications for targeted therapies?

CD46 is significantly overexpressed in multiple cancer types compared to normal cells, creating an opportunity for targeted therapeutic approaches:

Expression Level Comparison:

  • In multiple myeloma (MM) cell lines, CD46 antigen density ranges from 454,668 to 470,991 molecules per cell

  • This exceeds even CD38 (a common MM marker) which ranges from 314,953 to 344,865 molecules per cell

Genetic Basis for Overexpression:

  • The CD46 gene resides on chromosome 1q, which undergoes genomic amplification in the majority of relapsed myeloma patients

  • Patients with 1q gain show markedly higher cell surface expression of CD46 than those with normal 1q copy number

Therapeutic Implications:

  • Differential expression provides a therapeutic window for targeted approaches

  • The high density of CD46 on tumor cells facilitates efficient binding of therapeutic antibodies

  • Genomic amplification of CD46 may serve as a biomarker for patient stratification

This expression profile makes CD46 particularly attractive for antibody-drug conjugates and radioimmunotherapy approaches that require sufficient target density to deliver therapeutic payloads effectively .

What mechanisms drive the internalization of CD46 antibodies, and how does this impact therapeutic applications?

The internalization of CD46 antibodies is crucial for their therapeutic efficacy, particularly for antibody-drug conjugates (ADCs):

Internalization Mechanisms:

  • Some anti-CD46 antibodies are internalized via macropinocytosis, a tumor-selective pathway for cellular entry

  • Internalized antibodies colocalize with lysosomal-associated membrane protein 1 (LAMP1), indicating trafficking to lysosomes where ADC payloads can be released

Experimental Validation:

  • Confocal microscopy studies with MM1.R and RPMI8226 cells demonstrate that anti-CD46 antibody (23AG2) is effectively internalized and colocalizes with LAMP1

  • Binding kinetics measurements show KD values of 1.19 nM for RPMI8226 and 2.24 nM for MM1.S cells, indicating high-affinity binding that facilitates efficient internalization

Therapeutic Impact:

  • Efficient internalization enables delivery of cytotoxic payloads directly into tumor cells

  • Selective internalization in cancer cells contributes to the therapeutic window by sparing normal tissues

  • ADCs developed from internalizing anti-CD46 antibodies show potent cytotoxicity with EC50 values in the picomolar range

This internalization property has led to the development of various CD46-targeted therapeutics, including an ADC currently in clinical trials (NCT03575819) for metastatic castration-resistant prostate cancer .

How can researchers accurately quantify CD46 expression levels for experimental and clinical applications?

Accurate quantification of CD46 expression is essential for both research and potential patient stratification:

Flow Cytometry Quantification:

  • Flow cytometry using calibrated standards can determine absolute numbers of CD46 molecules per cell

  • This approach has revealed antigen densities of 454,668-470,991 molecules per cell in MM cell lines

Transcript Analysis:

  • CD46 transcript levels correlate inversely with the EC50 of CD46-ADC, suggesting mRNA quantification could predict therapeutic response

  • RT-PCR or RNA-seq can provide relative expression levels across samples

Immunohistochemistry Scoring:

  • Semi-quantitative scoring of IHC staining intensity (0-3+) can stratify expression levels in tissue samples

  • Recommended dilutions of 1:500-1:2000 for IHC applications with appropriate antigen retrieval methods

Western Blot Densitometry:

  • Quantitative Western blot with appropriate loading controls and standard curves can measure relative protein expression

  • Recommended dilutions of 1:2000-1:14000 for WB applications

Genetic Analysis:

  • FISH or other genetic assays to detect chromosome 1q amplification can serve as a surrogate marker for CD46 overexpression in certain cancers

For clinical applications, standardized assays with validated cutoff values would need to be established to categorize patients as "high" versus "low" CD46 expressors.

What are the critical parameters for optimizing antibody dilution and incubation conditions across different CD46 detection methods?

Optimization of antibody usage requires careful consideration of several parameters:

Recommended Dilution Ranges by Application:

ApplicationRecommended DilutionIncubation Parameters
Western Blot (WB)1:2000-1:14000Typically overnight at 4°C or 1-2 hours at room temperature
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg proteinOvernight incubation at 4°C with rotation
Immunohistochemistry (IHC)1:500-1:200030-60 minutes at room temperature or overnight at 4°C
Immunofluorescence (IF)/ICC1:50-1:5001-2 hours at room temperature or overnight at 4°C

Optimization Strategy:

  • Start with the middle of the recommended dilution range

  • Perform a dilution series spanning above and below this starting point

  • Assess signal-to-noise ratio, not just signal intensity

  • Use positive and negative controls to determine optimal conditions

  • Consider tissue/cell-specific factors that may necessitate adjustment

Additional Considerations:

  • Sample-dependent factors may require further titration in each specific testing system

  • For IHC, antigen retrieval method significantly impacts staining and should be optimized alongside antibody dilution

  • Temperature and duration of incubation can be adjusted to enhance sensitivity or reduce background

It is strongly recommended that each laboratory determine optimal conditions for their specific experimental system rather than relying solely on published parameters .

What controls are essential when validating CD46 antibody specificity and performance?

A robust validation strategy for CD46 antibodies should include multiple complementary controls:

Positive Expression Controls:

  • Cell lines: A549, HeLa, Jurkat, and MOLT-4 cells for Western blot and IF applications

  • Tissues: Human ovary tumor, breast cancer, colon cancer, or cervical cancer tissues for IHC applications

Negative Controls:

  • CD46 knockdown/knockout samples: Published applications using CD46 KD/KO are documented and should be included where possible

  • Isotype controls: Use same host species and isotype as the CD46 antibody but with no specific target

  • Secondary antibody-only controls: To assess non-specific binding of the detection system

Specificity Controls:

  • Peptide competition assays: Pre-incubating antibody with immunizing peptide should abolish specific signal

  • Multiple antibody approach: Use antibodies targeting different CD46 epitopes to confirm consistent detection

  • Validation across multiple applications: Concordant results across WB, IHC, and IF increase confidence in specificity

Technical Controls:

  • Loading controls: For quantitative Western blot applications

  • Tissue processing controls: For IHC to ensure consistent fixation and processing

  • For therapeutic applications (ADCs): Include isotype control ADCs to distinguish between target-specific and non-specific effects

Antibody Performance Metrics:

  • Binding kinetics: KD values (e.g., 2.99 nM on recombinant protein, 1.19-2.24 nM on living cells)

  • Limit of detection: Determine minimum detectable CD46 expression level

  • Dynamic range: Establish the range of expression levels that can be reliably distinguished

What methodological approaches are recommended for investigating CD46 regulation and function in experimental models?

Several complementary approaches can be employed to study CD46 regulation and function:

Gene Expression Manipulation:

  • CRISPR/Cas9 knockout: Complete elimination of CD46 to assess functional consequences

  • siRNA/shRNA knockdown: Partial reduction of expression for dose-dependent studies

  • Overexpression systems: Transfection with CD46 expression vectors for gain-of-function studies

  • Isoform-specific manipulation: Selective expression of individual CD46 isoforms

Functional Assays:

  • Complement regulation: Measure C3b/C4b deposition and complement-mediated lysis in CD46-manipulated cells

  • Receptor function: Assess measles virus binding and entry in models with altered CD46 expression

  • Cellular internalization: Track antibody internalization using fluorescently-labeled anti-CD46 antibodies

  • Signaling pathway analysis: Evaluate downstream effects of CD46 engagement on cellular signaling

Co-culture Systems:

  • Tumor-stromal interactions: CD46 expression can be upregulated upon co-culture with bone marrow stromal cells, suggesting microenvironmental regulation

  • Immune cell interactions: Investigate effects of CD46 on interactions with complement-producing cells

In Vivo Models:

  • Orthotopic xenograft models: Evaluate CD46 function in relevant tissue environments

  • Patient-derived xenografts: More closely recapitulate human disease complexity

  • Therapeutic targeting models: Test CD46-targeted therapies in preclinical models

When designing these studies, researchers should consider both the membrane-bound form of CD46 and potential shed forms, as shedding of extracellular CD46 antigen has been reported in some tumor cell lines .

What are the key design considerations for developing effective CD46-targeted antibody-drug conjugates?

The development of CD46-targeted ADCs requires optimization of multiple components:

Antibody Selection Criteria:

  • High specificity and affinity for CD46 (e.g., KD values in the low nanomolar range)

  • Efficient internalization properties, preferably via tumor-selective pathways like macropinocytosis

  • Binding to epitopes that are accessible in the tumor microenvironment

  • Minimal cross-reactivity with normal tissues

Linker-Payload Considerations:

  • Cleavable linkers: The valine-citrulline linker has shown success in CD46-ADCs, allowing lysosomal protease-dependent release

  • Payload selection: Monomethyl auristatin F (MMAF) conjugated to anti-CD46 antibodies has demonstrated potent cytotoxicity (EC50 in picomolar range)

  • Drug-antibody ratio (DAR): An average DAR of 3.3 has been effective in preclinical models

Efficacy and Safety Parameters:

  • Differential cytotoxicity: CD46-ADCs should show high potency against tumor cells (EC50 150 pM to 5 nM for MM cell lines) with minimal effect on normal cells (EC50 >100 nM for bone marrow stromal cells)

  • Mechanism of action: Confirm induction of apoptosis and cell death in target cells

  • Target expression correlation: Higher CD46 expression levels correlate with increased ADC potency

Current clinical development includes a CD46-targeted ADC in a multi-center phase I trial for metastatic castration-resistant prostate cancer (NCT03575819) , demonstrating the translational potential of this approach.

How does CD46-targeted radioimmunotherapy compare with antibody-drug conjugates in terms of mechanism and application?

CD46-targeted radioimmunotherapy and ADCs represent complementary approaches with distinct characteristics:

Radioimmunotherapy Characteristics:

  • Mechanism: Delivers ionizing radiation to tumor cells and surrounding microenvironment

  • Payload: Alpha-emitting radionuclides like 212Pb offer high linear energy transfer over short distances

  • Bystander effect: Can eliminate nearby tumor cells even if they lack CD46 expression

  • Example: 212Pb-TCMC-YS5 has shown potent anti-tumor activity in multiple prostate cancer models

ADC Characteristics:

  • Mechanism: Delivers cytotoxic agents directly into tumor cells following internalization

  • Payload: Typically microtubule inhibitors like MMAF that disrupt cell division

  • Cell-autonomous effect: Primarily affects cells that express CD46 and internalize the ADC

  • Example: CD46-ADC with MMAF shows picomolar potency against MM cell lines

Comparative Considerations:

  • Tumor penetration: Radioimmunotherapy may have advantages for poorly vascularized tumors

  • Heterogeneity: Radioimmunotherapy's bystander effect may better address tumor heterogeneity

  • Internalization requirements: ADCs require efficient internalization, while some radioimmunoconjugates can be effective without internalization

  • Safety profile: Different off-target toxicity considerations between radiation exposure and cytotoxic drug release

Both approaches have shown promise in preclinical models, with the 212Pb-TCMC-YS5 radioimmunotherapy being well-tolerated and showing potent anti-tumor activity in multiple prostate cancer models , while CD46-ADC demonstrated picomolar EC50 values against multiple myeloma cell lines with minimal effect on normal cells .

How can chromosome 1q amplification status be utilized as a biomarker for patient stratification in CD46-targeted therapies?

The relationship between chromosome 1q amplification and CD46 expression offers potential for patient stratification:

Biological Basis:

  • The CD46 gene resides on chromosome 1q, which undergoes genomic amplification in many cancers

  • In multiple myeloma, 1q gain occurs in the majority of relapsed patients

  • Patients with 1q gain show markedly higher cell surface expression of CD46 compared to those with normal 1q copy number

Implementation Strategy:

Biomarker ApproachMethodologyClinical Application
Direct 1q FISH analysisFluorescence in situ hybridization to detect 1q amplificationIdentify patients likely to have high CD46 expression
CD46 expression quantificationFlow cytometry on patient samples to measure CD46 antigen densityDirectly select patients with highest target expression
Combined genetic/expression analysisIntegrate 1q status with CD46 protein levelsRefine patient selection with multiple parameters

Clinical Implications:

  • Patients with 1q gain might benefit most from CD46-targeted therapies due to higher target expression

  • Genomic amplification serves as a surrogate marker for target amplification

  • The correlation between CD46 surface expression and ADC efficacy suggests expression-guided patient selection could improve outcomes

This approach represents a precision medicine strategy where genetic biomarkers (1q amplification) could identify patients most likely to benefit from CD46-directed therapies, potentially improving therapeutic index and clinical outcomes.

What methodological approaches are most effective for monitoring CD46-targeted therapy response in preclinical and clinical settings?

Comprehensive monitoring of CD46-targeted therapy requires multi-modal assessment approaches:

Preclinical Response Monitoring:

Clinical Response Evaluation:

  • RECIST criteria: Standard radiographic assessment of tumor response

  • Circulating tumor cells: Quantify CD46-expressing CTCs before and during treatment

  • Pharmacokinetic profiling: Measure drug exposure and clearance

  • Target occupancy: Assess CD46 occupancy on accessible tumor cells during treatment

  • Biomarker dynamics: Monitor changes in relevant biomarkers (e.g., M-protein for multiple myeloma)

Resistance Mechanism Identification:

  • CD46 expression changes: Monitor for potential downregulation during treatment

  • Epitope mutations: Sequence CD46 to detect binding-site alterations

  • Internalization defects: Assess changes in antibody internalization efficiency

  • Efflux pump upregulation: For ADCs, evaluate expression of drug efflux transporters

In both preclinical and clinical settings, correlating CD46 expression levels with therapeutic response provides valuable insights into the relationship between target density and efficacy, potentially allowing for refined patient selection and treatment optimization .

How can researchers address discrepancies between expected and observed molecular weights for CD46 in Western blot applications?

Discrepancies between calculated (43 kDa) and observed (typically 50-56 kDa, but ranging from 45-70 kDa) molecular weights of CD46 can be systematically addressed:

Common Causes and Solutions:

  • Post-translational modifications:

    • CD46 undergoes extensive glycosylation

    • Solution: Treat samples with glycosidases (PNGase F for N-linked glycans, O-glycosidase for O-linked glycans) prior to SDS-PAGE to reduce apparent molecular weight

  • Isoform variation:

    • Four predominant isoforms arise from alternative splicing

    • Solution: Use isoform-specific antibodies or primers to distinguish between variants

  • Sample preparation:

    • Reduction/denaturation conditions affect migration

    • Solution: Use non-reduced sample treatment for SDS-PAGE as recommended for some CD46 antibodies

  • Technical considerations:

    • Gel percentage, running buffer, and voltage affect migration

    • Solution: Include molecular weight standards appropriate for your gel system and standardize electrophoresis conditions

Verification Approaches:

  • Use multiple antibodies targeting different CD46 epitopes to confirm the identity of observed bands

  • Include positive control lysates from cells known to express CD46 (A549, HeLa, Jurkat, MOLT-4)

  • Consider mass spectrometry to definitively identify the protein and characterize its modifications

When reporting results, researchers should specify the observed molecular weight range and acknowledge these potential sources of variation to facilitate interpretation and comparison across studies.

What strategies can mitigate non-specific binding and optimize signal-to-noise ratio when using CD46 antibodies?

Optimizing signal-to-noise ratio requires addressing several potential sources of non-specific binding:

Blocking Optimization:

  • Test different blocking agents (BSA, non-fat dry milk, normal serum from secondary antibody species)

  • Optimize blocking duration and temperature

  • Consider specialized blocking reagents for problematic samples

Antibody Dilution Refinement:

  • Perform careful titration experiments to identify optimal concentration

  • For Western blot, dilutions of 1:2000-1:14000 are recommended

  • For IHC, dilutions of 1:500-1:2000 are recommended

  • For IF/ICC, dilutions of 1:50-1:500 are recommended

Washing Protocol Enhancement:

  • Increase number of washes

  • Extend washing duration

  • Add low concentrations of detergents (0.05-0.1% Tween-20) to wash buffers

  • Use agitation during washing steps

Sample-Specific Adjustments:

  • For IHC, optimize antigen retrieval methods (TE buffer pH 9.0 or citrate buffer pH 6.0)

  • For fixed samples, ensure fixation is not excessive

  • For Western blots, consider using freshly prepared samples

Advanced Approaches:

  • Pre-adsorb antibodies with tissues/cells lacking CD46 to remove cross-reactive antibodies

  • Use monovalent Fab fragments to reduce non-specific Fc-mediated binding

  • Consider using directly labeled primary antibodies to eliminate secondary antibody cross-reactivity

Importantly, each laboratory should determine the optimal conditions for their specific experimental system, as performance can be sample-dependent .

What approaches can address sample-specific variability in CD46 detection across different tissue types?

Tissue-specific variability in CD46 detection requires tailored methodological approaches:

Tissue-Specific Optimization Strategies:

  • Antigen Retrieval Customization:

    • Primary approach: TE buffer at pH 9.0

    • Alternative approach: Citrate buffer at pH 6.0

    • Optimize duration and temperature for each tissue type

    • Consider microwave vs. pressure cooker methods based on tissue density

  • Fixation Considerations:

    • Duration of fixation significantly impacts epitope preservation

    • Overfixation may mask epitopes, while inadequate fixation compromises morphology

    • Consider testing different fixatives for challenging tissues

  • Antibody Selection by Tissue:

    • Different anti-CD46 antibodies may perform better in specific tissue contexts

    • Test multiple antibodies targeting different CD46 epitopes

    • Consider potential tissue-specific post-translational modifications affecting epitope accessibility

  • Signal Amplification Methods:

    • For tissues with lower CD46 expression, employ tyramide signal amplification or other amplification systems

    • Balance amplification with potential increases in background

Standardization Approaches:

  • Include universal positive control tissues (e.g., placenta or prostate) alongside test samples

  • Develop tissue-specific protocols based on systematic optimization

  • Consider multiplexed detection with invariant markers to normalize CD46 signal

Technical Validation:

  • Confirm CD46 detection using orthogonal methods (e.g., RNAscope for mRNA detection)

  • Use automated staining platforms to minimize technical variability when possible

  • Implement digital image analysis for objective quantification across tissue types

The search results indicate successful detection of CD46 in various cancer tissues (ovary, breast, colon, and cervical) , suggesting that with proper optimization, reliable CD46 detection is achievable across diverse tissue types.

Product Science Overview

Introduction

CD46, also known as Membrane Cofactor Protein (MCP), is a type I membrane glycoprotein that plays a crucial role in the regulation of the complement system. It is ubiquitously expressed on all nucleated cells and serves as a cofactor for the serine protease factor I, which mediates the inactivation of C3b and C4b deposited on host cells . This function is vital in preventing complement-mediated damage to host tissues.

Discovery and Function

CD46 was first discovered in 1986 during a search for novel C3b-binding proteins . Its primary function is to protect host cells from complement-mediated injury by facilitating the cleavage of C3b and C4b into inactive fragments. This process is essential for maintaining self-tolerance and preventing autoimmune reactions.

In addition to its role in complement regulation, CD46 has been found to participate in various other biological processes, including the modulation of adaptive immune responses. It is involved in the regulation of T helper type 1 (Th1) immune responses by influencing the production of interferon-gamma (IFN-γ) and interleukin-10 (IL-10) within these cells . This dual role in both innate and adaptive immunity underscores the importance of CD46 in maintaining immune homeostasis.

Clinical Significance

Deficiency or dysfunction of CD46 is associated with several disease conditions. For instance, mutations in the CD46 gene can predispose individuals to atypical hemolytic uremic syndrome (aHUS), a rare but severe condition characterized by the destruction of red blood cells, low platelet count, and kidney failure . Additionally, CD46 has been implicated in various autoimmune diseases and infections, as many pathogens exploit CD46 as a receptor to gain entry into host cells .

Mouse Anti-Human CD46 Antibodies

Mouse anti-human CD46 antibodies are widely used in research and clinical diagnostics to study the expression and function of CD46. These antibodies are typically generated by immunizing mice with human CD46 protein, followed by the isolation and purification of the resulting antibodies. They are available in various formats, including unconjugated and fluorochrome-conjugated forms, to facilitate different experimental applications such as flow cytometry and immunohistochemistry .

Applications in Research

Mouse anti-human CD46 antibodies are valuable tools in immunological research. They are used to investigate the role of CD46 in immune regulation, complement activation, and disease pathogenesis. For example, flow cytometric analysis using these antibodies can help quantify CD46 expression on different cell types and assess its involvement in various immune responses . Additionally, these antibodies can be used to study the mechanisms by which pathogens interact with CD46 and develop strategies to block these interactions for therapeutic purposes.

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.