CHRND Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery times may vary depending on the shipping method and location. For specific delivery timeframes, please consult your local distributor.
Synonyms
Acetylcholine receptor delta subunit antibody; Acetylcholine receptor subunit delta antibody; ACHD_HUMAN antibody; ACHRD antibody; Cholinergic receptor, nicotinic, delta polypeptide antibody; CHRND antibody; CMS2A antibody; FCCMS antibody; Nicotinic acetylcholine receptor delta polypeptide precursor antibody; SCCMS antibody
Target Names
CHRND
Uniprot No.

Target Background

Function
Upon binding acetylcholine, the AChR undergoes a significant conformational change affecting all subunits. This alteration results in the opening of an ion-conducting channel across the plasma membrane.
Gene References Into Functions
  1. Research suggests that a mutation in the invariant Cys-loop of CHRND (D140N) observed in the muscle of a patient (11-year-old girl) with congenital myasthenia (characterized by severe muscle weakness) alters the conformation of the ligand/acetylcholine binding site and receptor functionality. PMID: 26698174
  2. This study demonstrated that a single mutation in the delta subunit, L332P, allows the synapse in slow muscles to function while rendering those in fast muscles almost nonfunctional. PMID: 25080583
  3. This research describes the effects of a point mutation in the AChR delta subunit from a congenital myasthenia patient. PMID: 18398509
Database Links

HGNC: 1965

OMIM: 100720

KEGG: hsa:1144

STRING: 9606.ENSP00000258385

UniGene: Hs.156289

Involvement In Disease
Multiple pterygium syndrome, lethal type (LMPS); Myasthenic syndrome, congenital, 3A, slow-channel (CMS3A); Myasthenic syndrome, congenital, 3B, fast-channel (CMS3B); Myasthenic syndrome, congenital, 3C, associated with acetylcholine receptor deficiency (CMS3C)
Protein Families
Ligand-gated ion channel (TC 1.A.9) family, Acetylcholine receptor (TC 1.A.9.1) subfamily, Delta/CHRND sub-subfamily
Subcellular Location
Cell junction, synapse, postsynaptic cell membrane; Multi-pass membrane protein. Cell membrane; Multi-pass membrane protein.

Q&A

What is CHRND antibody and why is biotin conjugation significant for research applications?

CHRND antibody targets the delta subunit of the nicotinic acetylcholine receptor (AChR), a crucial component in neuromuscular junction signaling. The delta subunit (Cholinergic Receptor, Nicotinic, delta Muscle) plays an essential role in receptor assembly and function. After acetylcholine binding, AChR undergoes extensive conformational changes affecting all subunits, leading to the opening of an ion-conducting channel across the plasma membrane .

Biotin conjugation significantly enhances the utility of CHRND antibodies by enabling:

  • Increased sensitivity in detection systems through biotin-streptavidin amplification

  • Multi-layered staining protocols without cross-reactivity issues

  • Flexibility in experimental design with various secondary detection systems

  • Enhanced signal-to-noise ratio in complex tissue samples

Multiple commercial biotin-conjugated CHRND antibodies are available, with applications spanning Western blotting, ELISA, flow cytometry, and immunofluorescence . This conjugation creates a versatile research tool particularly valuable for studying receptor clustering and trafficking in neuromuscular junction disorders.

How do I select the appropriate CHRND antibody epitope region for myasthenia gravis research?

Selection of the appropriate epitope region is critical when investigating myasthenia gravis pathophysiology. Research demonstrates that antibodies targeting different domains of the CHRND protein can produce varying experimental outcomes:

  • N-terminal region antibodies (AA 22-245, AA 27-76) are optimal for studying receptor assembly and ligand binding interfaces

  • Mid-region antibodies (AA 168-196) better detect structural changes during channel activation

  • C-terminal region antibodies (AA 334-471) are preferred for investigating cytoplasmic signaling interactions

For myasthenia gravis research specifically, antibodies recognizing the extracellular domain (AA 22-245) have demonstrated superior performance in cell-based assays (CBAs). These antibodies can effectively detect pathogenic autoantibodies that target similar regions in patient samples .

When selecting an antibody, evaluate:

  • The specific research question (receptor assembly vs. clustering vs. internalization)

  • Required species cross-reactivity (human, rat, etc.)

  • Intended applications (flow cytometry vs. Western blotting)

  • Compatibility with your experimental system (cell lines, tissue samples)

The antibody clone 1H1F9 has been documented with successful application in myasthenia gravis research, particularly in flow cytometric analysis of neuroblastoma cell lines .

What experimental controls should be implemented when using biotin-conjugated CHRND antibodies?

Implementing appropriate controls is essential for ensuring result validity when working with biotin-conjugated CHRND antibodies:

Essential Control Panel:

Control TypePurposeImplementation
Isotype ControlAssesses non-specific bindingUse biotin-conjugated mouse IgG1 (for monoclonal) or species-matched IgG (for polyclonal) at identical concentrations
Blocking ControlConfirms binding specificityPre-incubate antibody with recombinant CHRND protein (AA 22-245)
Endogenous Biotin ControlEvaluates endogenous biotin interferenceInclude avidin/streptavidin-only stained samples
Cell Line ValidationVerifies target expressionCompare CHRND-expressing (e.g., SK-N-SH, C6) with non-expressing cell lines
Secondary Reagent ControlDetects background from detection systemInclude samples with secondary reagents only

For cell-based assays, transfect HEK293T cells with adult (2α, β, δ, and ε) or fetal (2α, β, δ, and γ) AChR subunits along with rapsyn-GFP to create specialized positive controls . This approach enables confirmation of specific subunit recognition within the assembled receptor complex.

Additionally, when working with patient samples in myasthenia gravis research, include control samples from healthy donors and non-myasthenic neurological disease patients to establish appropriate baseline measurements and specificity parameters .

How can biotin-conjugated CHRND antibodies be optimized for cell-based assays in myasthenia gravis research?

Optimizing biotin-conjugated CHRND antibodies for cell-based assays in myasthenia gravis research requires sophisticated methodological considerations:

  • Expression System Engineering:

    • Transiently transfect HEK293T cells with precise ratios of AChR subunits (2α, β, δ, and ε for adult; 2α, β, δ, and γ for fetal) along with rapsyn-GFP using branched polyethylenimine

    • Maintain transfected cells for 48 hours before harvesting, with medium replenishment at 24 hours to maximize receptor expression

  • Detection Protocol Refinement:

    • Incubate AChR-expressing cells with biotin-conjugated CHRND antibodies for 1 hour at 4°C to minimize receptor internalization

    • After washing, apply streptavidin-conjugated fluorophores with spectral properties compatible with rapsyn-GFP visualization

    • For multiplex assays, confirm appropriate CHRND subunit expression with complementary antibodies: anti-alpha (mAb35), anti-CHRNB1, anti-CHRNE, and anti-CHRNG

  • Quantification Strategy:

    • Employ flow cytometry for objective quantification of binding using mean fluorescence intensity

    • Establish a standardized gating strategy based on rapsyn-GFP expression level to normalize receptor density variations

    • Calculate binding indices as ratios compared to reference standards for consistent inter-assay comparability

Recent comparative studies demonstrate that cell-based assays utilizing biotin-conjugated antibodies exhibit superior sensitivity (72.3%, 95% CI: 70.3–74.3) compared to radioimmunoprecipitation assays (64.1%, 95% CI: 62.0–66.2) and ELISA (62.7%, 95% CI: 60.5–64.8) for detecting AChR autoantibodies in myasthenia gravis patients .

What methodological approaches can distinguish between pathogenic and non-pathogenic antibody binding to CHRND in autoimmune disorders?

Distinguishing pathogenic from non-pathogenic antibody binding requires advanced functional assays that assess the biological consequences of CHRND antibody interaction:

  • Complement Activation Assessment:

    • Utilize modified triple-knockout HEK293T cells (devoid of CD46, CD55, CD59 complement regulator genes) expressing AChR

    • Incubate cells with test antibodies in the presence of complement-competent normal human serum for 2 hours at 37°C

    • Detect antibody-dependent membrane attack complex (MAC) formation using anti-C9 neoantigen antibody followed by fluorescently-labeled secondary antibodies

    • Quantify complement activation as an indicator of pathogenicity

  • Receptor Modulation Assay:

    • Culture human rhabdomyosarcoma CN21 muscle cells expressing endogenous AChR

    • Pre-incubate with purified antibodies (1 μg/mL) for 16 hours at 37°C

    • Quantify remaining surface AChR using fluorophore-conjugated α-bungarotoxin

    • Calculate the modulation index as the percentage of receptor internalization relative to controls

    • Pathogenic antibodies typically induce >20% receptor modulation

  • Functional Electrophysiology:

    • Patch-clamp recordings of AChR-expressing cells before and after antibody application

    • Measure changes in acetylcholine-evoked current amplitude and kinetics

    • Analyze alterations in single-channel conductance properties

    • Pathogenic antibodies often cause functional channel blockade or altered opening probability

Research has demonstrated that biotin-conjugated CHRND antibodies that induce receptor internalization and complement activation correlate strongly with clinical disease severity in myasthenia gravis patients, providing valuable diagnostic and prognostic information .

How do modifications in CHRND antibody biotinylation density affect experimental outcomes in multiplex immunoassays?

The biotinylation density of CHRND antibodies significantly impacts experimental outcomes in multiplex immunoassays through several mechanisms:

Biotinylation Density Effects:

Biotinylation RatioSignal IntensityBackgroundCross-ReactivityReceptor Accessibility
Low (1-2 biotin/Ab)ModerateMinimalLowHigh
Medium (3-5 biotin/Ab)OptimalLowModerateModerate
High (>6 biotin/Ab)Initially high, potential signal quenchingIncreasedHigher riskReduced

For multiplex immunoassays specifically, maintaining a controlled biotinylation ratio (preferably 3-5 biotin molecules per antibody) provides optimal signal-to-noise ratios while preserving antibody functionality. Excessive biotinylation can create structural distortions that paradoxically reduce binding affinity to CHRND epitopes.

Advanced technical considerations include:

  • Validating each new batch of biotin-conjugated antibodies for consistency in biotinylation ratio

  • Implementing titration experiments to determine optimal concentrations for each application

  • Developing compensation matrices for spectral overlap when using multiple biotin-conjugated antibodies in the same assay

  • Considering steric interference when targeting closely positioned epitopes on assembled AChR complexes

Recent biotin supplementation studies provide insight into the relationship between biotin availability and protein interactions. Biotin supplementation at 10 μM represents a minimum effective concentration for enhancing detection sensitivity without introducing artifacts from excess free biotin .

What are the best methodological approaches for simultaneously detecting multiple AChR subunits including CHRND in complex tissue samples?

Simultaneous detection of multiple AChR subunits in complex tissue samples requires sophisticated methodological approaches:

  • Multiplexed Immunofluorescence Strategy:

    • Employ a sequential staining protocol with primary antibodies from different host species

    • For CHRND specifically, utilize biotin-conjugated antibodies followed by streptavidin-fluorophore detection

    • Include antibodies against alpha (mAb35), beta (anti-CHRNB1), epsilon (anti-CHRNE), and gamma (anti-CHRNG) subunits

    • Apply spectral unmixing algorithms to resolve overlapping emission spectra

    • Implement tissue clearing techniques (CLARITY, iDISCO) for improved antibody penetration in thick tissue sections

  • Proximity Ligation Assay (PLA):

    • Utilize paired antibodies against adjacent subunits (e.g., CHRND and CHRNA1)

    • Apply secondary antibodies with attached DNA oligonucleotides

    • Generate fluorescent signals only when target proteins are in close proximity (<40 nm)

    • This approach verifies assembled receptor complexes rather than individual subunits

  • Cell-Based Assay Adaptation for Tissue Analysis:

    • Apply fixed CBA methodology similar to clinical diagnostic approaches

    • Transfect HEK293T cells with α, β, δ, γ and ε subunits in a 2:1:1:1:1 ratio respectively

    • Fix cells with 4% polyformaldehyde after transfection

    • Incubate with tissue extracts or serum samples

    • Detect bound antibodies using fluorescently-labeled secondary antibodies

For validating specificity in tissue samples, confirm CHRND detection using antibodies targeting different epitopes: N-terminal (AA 27-76), mid-region (AA 168-196), and C-terminal (AA 334-471) domains .

How does biotin supplementation affect experimental outcomes when using biotin-conjugated CHRND antibodies?

Biotin supplementation during experiments with biotin-conjugated CHRND antibodies requires careful consideration due to several potential interference mechanisms:

  • Competition for Streptavidin Binding:

    • Excessive free biotin from supplementation can compete with biotin-conjugated antibodies for streptavidin binding sites

    • This competition reduces signal intensity in a dose-dependent manner

    • At biotin concentrations >10 μM, significant signal reduction occurs in most detection systems

  • Altered Histone Biotinylation and Gene Expression:

    • Biotin supplementation (10 μM) significantly increases histone biotinylation at H1, H2A, H2B, H3, and H4

    • This epigenetic modification can alter gene expression patterns, potentially including AChR subunit genes

    • Increased histone biotinylation has been observed to colocalize with Xist signal, suggesting effects on chromosome inactivation

    • These changes may alter the baseline expression of target proteins in experimental systems

  • Metabolic Effects on Cell Models:

    • Biotin supplementation influences autophagy markers, including increased LC3-II levels

    • This metabolic shift may alter membrane protein trafficking, including AChR surface expression

    • These effects become significant in experiments examining receptor internalization or turnover

Recommended Mitigation Strategies:

  • For short-term experiments (<24 hours), avoid biotin supplementation in culture media

  • For long-term studies, establish baseline measurements of receptor expression under standard and biotin-supplemented conditions

  • Include avidin blocking steps in staining protocols when working with biotin-supplemented samples

  • Consider using alternative conjugation chemistries (e.g., Alexa Fluor direct conjugation) for experiments involving biotin supplementation

  • If biotin supplementation is necessary, implement validation experiments to determine the minimum effective concentration (typically 1-10 μM)

What are the optimal fixation and permeabilization protocols for preserving CHRND epitope integrity in immunohistochemistry?

Preserving CHRND epitope integrity requires careful optimization of fixation and permeabilization protocols, particularly due to the complex tertiary structure of the receptor:

Fixation Protocol Comparison:

Fixation MethodCHRND Epitope PreservationTissue MorphologyBackgroundRecommended Application
4% Paraformaldehyde (15 min, 4°C)Excellent for extracellular domainsGoodLowFixed cell-based assays, tissue sections
2% Paraformaldehyde (10 min, RT)Very good for most domainsModerateVery lowFlow cytometry, cultured cells
Methanol (-20°C, 10 min)Good for intracellular domainsFairModerateIntracellular epitope access
Acetone (4°C, 10 min)Variable by epitope regionPoorHigherFrozen sections only
Glutaraldehyde (0.1%, 5 min)Poor for most CHRND epitopesExcellentHighNot recommended

For biotin-conjugated CHRND antibodies specifically, mild fixation with 2-4% paraformaldehyde for limited duration provides optimal results. This preserves epitope structure while maintaining tissue architecture.

Permeabilization Optimization:

  • For extracellular domains (AA 22-245): Avoid permeabilization entirely

  • For transmembrane regions: Mild detergents (0.1% Triton X-100, 5 minutes)

  • For intracellular domains (AA 334-471): 0.2-0.3% Triton X-100 or 0.1% saponin

To maximize detection of assembled AChR complexes in tissue sections, implement an antigen retrieval step using citrate buffer (pH 6.0) at 95°C for 15-20 minutes, followed by a cooling period of 20 minutes before antibody application.

How can biotinylated CHRND antibodies be effectively utilized in super-resolution microscopy of neuromuscular junctions?

Utilizing biotinylated CHRND antibodies in super-resolution microscopy requires specialized approaches to overcome resolution limitations and maximize signal quality:

  • Sample Preparation Optimization:

    • Fresh tissue fixation with 2% paraformaldehyde (no methanol)

    • Careful sectioning to minimize out-of-plane fluorescence

    • Implementation of optical clearing techniques (SeeDB, ScaleS) to improve imaging depth

    • Use of ultrathin (70-100 nm) sections for Structured Illumination Microscopy (SIM)

  • Detection Strategy Enhancement:

    • Apply biotin-conjugated primary anti-CHRND antibodies at optimized concentration (typically 1-5 μg/mL)

    • Detect with streptavidin conjugated to specialized super-resolution compatible fluorophores:

      • Alexa Fluor 647 for STORM/PALM

      • Atto 488 for STED

      • SiR-based dyes for live-cell super-resolution

    • Consider implementing DNA-PAINT methodology for highest resolution:

      • Conjugate DNA oligonucleotides to streptavidin

      • Use complementary fluorophore-labeled DNA strands for transient binding

  • Multicolor Imaging Strategy:

    • When visualizing CHRND in relation to other subunits or proteins:

      • Use biotin-conjugated CHRND antibodies with streptavidin-conjugated far-red fluorophores

      • Apply directly conjugated antibodies against other targets in compatible channels

      • Include rapsyn visualization (via GFP fusion) to identify clustered receptors

    • For presynaptic/postsynaptic differentiation:

      • Pair CHRND detection with synaptophysin or bassoon markers

      • Implement channel alignment strategies using fiducial markers

Super-resolution techniques have revealed that CHRND subunits exhibit specific nanoscale distribution patterns within AChR clusters. STORM imaging specifically has demonstrated that these patterns differ between healthy neuromuscular junctions and those affected by autoimmune pathologies, providing valuable insights into disease mechanisms.

How do cell-based assays using biotin-conjugated CHRND antibodies compare to radioimmunoprecipitation assays in myasthenia gravis diagnostics?

Cell-based assays (CBAs) using biotin-conjugated CHRND antibodies offer several distinct advantages over traditional radioimmunoprecipitation assays (RIPAs) in myasthenia gravis diagnostics:

Performance Comparison:

ParameterCell-Based Assay (CBA)Radioimmunoprecipitation Assay (RIPA)Enzyme-Linked Immunosorbent Assay (ELISA)
Sensitivity72.3% (95% CI: 70.3–74.3)64.1% (95% CI: 62.0–66.2)62.7% (95% CI: 60.5–64.8)
Specificity97.8% (95% CI: 95.0–99.3)97.8% (95% CI: 95.0–99.3)94.8% (95% CI: 91.9–97.7)
Detection of Low-Affinity AntibodiesExcellentPoorModerate
Conformational Epitope DetectionPreservedPartially disruptedDisrupted
Radiation Safety RequirementsNoneSignificantNone
Turnaround Time1-2 days2-3 days1 day

In a prospective multicenter study involving 2,272 participants (2,043 MG patients and 229 controls), CBAs detected AChR antibodies in 1,478 MG patients compared to 1,310 with RIPA and 1,280 with ELISA . This represents an absolute improvement in detection yield of 8.2-9.6%.

Methodological Advantages of CBA:

  • Preserves native protein conformation by expressing AChR subunits in mammalian cells

  • Enables visualization of receptor clustering when co-expressed with rapsyn-GFP

  • Allows for discrimination between adult and fetal AChR autoantibodies

  • Provides a non-radioactive alternative with improved safety profile

  • Facilitates multiplex analysis when combined with other fluorescent markers

For research applications specifically, biotin-conjugated CHRND antibodies in CBAs offer enhanced flexibility for experimental design and integration with complementary techniques such as flow cytometry and advanced microscopy.

What are the methodological differences between detecting adult versus fetal AChR with biotin-conjugated CHRND antibodies?

The methodological approach for detecting adult versus fetal AChR requires careful consideration of subunit composition and experimental design:

Key Methodological Differences:

ParameterAdult AChR DetectionFetal AChR Detection
Subunit Compositionα2βδεα2βδγ
Key Differential SubunitCHRNE (epsilon)CHRNG (gamma)
HEK293T Transfection Ratio2α:1β:1δ:1ε2α:1β:1δ:1γ
Antibody Validationanti-CHRNE (MA5-31698)anti-CHRNG (TA330432)
Predominant ExpressionAdult muscle, innervated NMJDeveloping muscle, denervated adult muscle
Disease RelevanceTypical myasthenia gravisFetal AChR syndrome, muscle denervation

While CHRND (delta subunit) is present in both receptor types, the context of detection requires different experimental approaches:

  • Cell-Based Assay Implementation:

    • For adult AChR: Transfect HEK293T cells with adult subunits (2α, β, δ, and ε)

    • For fetal AChR: Transfect with fetal subunits (2α, β, δ, and γ)

    • Include rapsyn-GFP in both systems to promote receptor clustering

  • Validation Strategy:

    • Confirm specific subunit expression using subtype-specific antibodies

    • Adult: anti-alpha (mAb35), anti-CHRNB1, anti-CHRND, anti-CHRNE

    • Fetal: anti-alpha (mAb35), anti-CHRNB1, anti-CHRND, anti-CHRNG

  • Specialized Applications:

    • For developmental studies: Track transition from fetal to adult AChR expression

    • For disease models: Examine reversion to fetal AChR expression after denervation

    • For autoimmune specificity: Determine if patient antibodies preferentially target adult vs. fetal receptors

The methodological distinction is particularly important in myasthenia gravis research, where antibodies with differential binding to adult versus fetal receptors may correlate with distinct clinical phenotypes and treatment responses .

How can high background signals be reduced when using biotin-conjugated CHRND antibodies in tissues with endogenous biotin?

High background signals from endogenous biotin represent a significant challenge when using biotin-conjugated antibodies. Implementing a comprehensive mitigation strategy is essential:

  • Endogenous Biotin Blocking Protocol:

    • Pre-treat tissue sections with avidin (10-50 μg/mL) for 15 minutes

    • Follow with biotin solution (50-200 μg/mL) for 15 minutes

    • Rinse thoroughly between steps and before applying primary antibody

    • For particularly biotin-rich tissues (liver, kidney), increase avidin concentration to 100 μg/mL

  • Alternative Detection Strategies:

    • For tissues with extremely high endogenous biotin:

      • Consider direct fluorophore conjugation of anti-CHRND antibodies

      • Utilize non-biotin amplification systems (e.g., polymer-based detection)

      • Implement tyramide signal amplification using HRP-conjugated secondaries

  • Sample Preparation Optimization:

    • Select fixation protocols that minimize biotin accessibility

    • Avoid prolonged fixation which can expose additional biotin epitopes

    • For frozen sections, implement shorter acetone fixation (5 minutes) which preserves antigenicity while reducing biotin accessibility

  • Antibody Selection and Validation:

    • Test different CHRND antibody clones for performance in biotin-rich tissues

    • The 1H1F9 clone shows lower non-specific binding in certain tissues

    • Compare performance of biotin-conjugated versus unconjugated CHRND antibodies in parallel sections

  • Advanced Signal Processing:

    • Implement computational background correction during image analysis

    • Use spectral unmixing to separate specific signal from autofluorescence

    • Apply localized background subtraction algorithms based on tissue morphology

Recent research on biotin supplementation has demonstrated that tissues from subjects with increased biotin intake exhibit significantly elevated background, requiring more rigorous blocking protocols . In tissues with naturally high biotin content, consider using alternative detection approaches entirely.

What strategies address inconsistent results from different lots of biotin-conjugated CHRND antibodies?

Addressing lot-to-lot variability in biotin-conjugated CHRND antibodies requires systematic validation and standardization strategies:

  • Comprehensive Lot Validation Protocol:

    • Establish a reference control system using well-characterized samples

    • For each new antibody lot, perform parallel testing against reference lot

    • Quantify critical parameters:

      • Signal intensity across a dilution series

      • Background levels in negative control tissues

      • Specificity using CHRND-expressing vs. non-expressing cells

      • Biotinylation ratio using HABA assay or mass spectrometry

  • Standardization Approaches:

    • Normalize antibody concentration based on biotin-to-protein ratio

    • Adjust working dilutions based on comparative titration curves

    • Implement internal calibration standards for quantitative applications

    • Document lot-specific optimal conditions in detailed protocol amendments

  • Technical Adaptations:

    • For flow cytometry: Adjust compensation matrices for each lot

    • For imaging: Standardize exposure settings using calibration slides

    • For Western blotting: Normalize loading and exposure times

    • For multiplex assays: Re-validate spectral overlap and detection thresholds

  • Supplier Engagement:

    • Request certificate of analysis including biotinylation ratio

    • Inquire about manufacturing changes that might affect conjugation

    • Consider purchasing larger lots for long-term studies

    • Engage supplier in troubleshooting persistent issues

Systematic evaluation of antibody lots should include testing against a panel of controls including:

  • Positive control (C6 rat glial tumor or SK-N-SH human neuroblastoma cell lines)

  • Negative control (cell lines not expressing CHRND)

  • Isotype control (biotinylated IgG1 for monoclonal antibodies)

By implementing these strategies, researchers can significantly reduce experimental variability and ensure consistent results across studies.

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.