17 kDa cell wall Antibody

Shipped with Ice Packs
In Stock

Description

17 kDa Proteins in Cellular Transport and Membrane Function

A 17 kDa proteolipid subunit of the vacuolar H⁺-ATPase (V-ATPase) has been identified in insect tissues, where it plays a role in proton conduction across apical membranes. This subunit is critical for ion regulation in midgut and Malpighian tubules.

Key Findings

SourceFunctionAntibody TypeApplicationReference
Heliothis virescensProton channel in V-ATPase complexRabbit polyclonalImmunolocalization, Western blot
Manduca sextaApical membrane localizationAnti-peptide antibodyNorthern blot, IHC

Mechanistic Insights

  • The 17 kDa subunit is part of the transmembrane sector of V-ATPase, facilitating proton transport.

  • Antibodies against this subunit label apical membranes in insect tissues, confirming its role in acid secretion and ion balance .

17 kDa Proteins in Immune Regulation

Several 17 kDa proteins are implicated in immune responses, including cytokines and antimicrobial peptides.

IL-17 Antibodies

IL-17A, a 17 kDa pro-inflammatory cytokine, is detected by specific monoclonal antibodies (e.g., MAB3171 and D1X7L).

AntibodyApplicationCross-ReactivityDetection MethodReference
MAB3171 (Mouse)ELISA, Western blot, IHCHuman IL-17A/IL-17FLymphocyte staining in Crohn’s disease
AF-317-NA (Goat)Western blot, IHCCanine IL-17 (~30%)Detection in Th17 cell lysates
D1X7L (Rabbit)Flow cytometry, IPMouse IL-17AEndogenous IL-17 detection

Functional Role

  • IL-17A drives inflammation in autoimmune diseases (e.g., Crohn’s disease) and is localized to lymphocytes in affected tissues .

  • Antibodies like MAB3171 enable precise quantification of IL-17A in ELISA and immunohistochemical assays .

Lactoferrin-Derived 17 kDa Fragment

A 17 kDa fragment of lactoferrin (Lf) promotes resolution of inflammation by enhancing macrophage efferocytosis and reducing pro-inflammatory cytokines.

FunctionMechanismApplicationReference
Anti-inflammatoryFKD/FKE tripeptides regulate macrophagesIn vivo peritonitis models
Pro-resolvingIncreases CD11b⁺ macrophagesBovine mastitis studies

Lettuce-Associated Allergen

A 17 kDa kirola-like protein in lettuce has been identified as a novel allergen in respiratory allergy cases.

AllergenDetection MethodPatient ReactivityReference
Kirola-like proteinImmunoblot, LC-MS/MS3/13 lettuce-allergic patients

Relevance

  • This protein represents a divergent allergen from previously identified lettuce antigens (e.g., EP1-like glycoprotein) .

Mycobacterium PstS1

While PstS1 itself is a 38 kDa phosphate transporter, antibodies targeting it (e.g., p4-163) inhibit Mycobacterium tuberculosis growth.

AntibodyTargetMechanismActivityReference
p4-163PstS1Blocks phosphate uptake~0.5 log reduction in bacterial load

17 kDa Prolactin Fragment

A 17 kDa N-terminal fragment of prolactin (PRLdel159), generated by BMP1 cleavage, exhibits antiangiogenic properties.

PropertyEffectModelReference
AntiangiogenicInhibits bFGF-induced endothelial proliferationHUVEC assays
ERK1/2 dephosphorylationReduces MAPK signalingPhosphorylation assays

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
17 kDa cell wall protein antibody; Fragment antibody
Uniprot No.

Target Background

Subcellular Location
Secreted, cell wall.

Q&A

What is a 17 kDa cell wall antibody and what cellular structures does it typically recognize?

17 kDa cell wall antibodies comprise a diverse group of immunological reagents that recognize specific epitopes on cell wall proteins or proteolipids with molecular weights of approximately 17 kilodaltons. These antibodies are primarily designed to detect structural components in various biological systems. In plant systems, they often target glycan epitopes present on cell wall components . In microbial systems, they may recognize specific proteolipids in membrane structures. For instance, a rabbit polyclonal antibody developed against a 17 kDa proteolipid successfully labeled the midgut goblet cell apical membrane in Heliothis virescens and the apical membrane in Malpighian tubules . In mammalian systems, such as human tissues, 17 kDa antibodies like the human IL-17/IL-17A antibody specifically bind to interleukins expressed by differentiated Th17 cells and can be detected in tissues like Crohn's disease intestine, where specific staining is localized to lymphocytes .

The methodology for confirming target specificity typically involves immunoblotting verification against purified protein preparations or cell lysates containing the target antigen. Researchers should always validate antibody specificity through Western blot analysis before proceeding to more complex applications.

How do I determine the optimal working dilution for a 17 kDa cell wall antibody in various applications?

Determining optimal working dilutions requires systematic titration experiments across multiple applications. Begin with manufacturer recommendations as baseline parameters, then perform dilution series experiments as follows:

  • For Western blot applications: Test a gradient of antibody concentrations (e.g., 0.1-5 μg/mL) against your target sample and a positive control. For instance, when detecting IL-17 in human samples, a concentration of 2 μg/mL has been successfully used with HRP-conjugated secondary antibodies .

  • For immunohistochemistry: A tiered approach starting with 0.5-2 μg/mL for paraffin-embedded tissues is recommended. In a documented case, human IL-17/IL-17A was successfully detected in fixed paraffin-embedded sections of Crohn's disease intestine using 0.5 μg/mL of the monoclonal antibody .

  • For flow cytometry: Start with 1-5 μg per 10^6 cells and adjust based on signal-to-noise ratio. Protocols often require fixation with paraformaldehyde and permeabilization with saponin to facilitate intracellular staining .

  • For immunoprecipitation: Typically requires higher concentrations, with successful results reported using 3 μg of antibody per 100 μg of cell lysate when incubated overnight at 4°C .

Document signal-to-noise ratios for each dilution and select the concentration that provides maximum specific signal with minimal background. Optimization should be performed for each new tissue type, species, or experimental condition.

What are the recommended storage and handling conditions to maintain antibody activity?

Proper storage and handling are critical for maintaining antibody performance over time. Based on empirical research data for 17 kDa antibodies, the following guidelines maximize stability and function:

For long-term storage:

  • Store at -20°C to -70°C in small aliquots to avoid repeated freeze-thaw cycles

  • Stability data suggests maintenance of activity for up to 12 months under these conditions

For working solutions:

  • After reconstitution, store at 2-8°C under sterile conditions for up to one month

  • For extended use, reconstituted antibodies can be stored at -20°C to -70°C for up to 6 months

Avoid:

  • Repeated freeze-thaw cycles (limit to <5)

  • Exposure to direct light

  • Storage in frost-free freezers with automatic defrost cycles

When handling, maintain sterile conditions and use appropriate buffer systems that match your experimental conditions. For specialized applications requiring buffer optimization, consult application-specific protocols, such as recommended buffer recipes for immunoprecipitation .

What are the most effective epitope retrieval methods for using 17 kDa cell wall antibodies in fixed tissue samples?

Epitope retrieval is critical when using 17 kDa cell wall antibodies on fixed tissues due to potential masking of epitopes during fixation. Based on empirical data, the following methods have proven most effective:

For paraffin-embedded sections:

  • Heat-induced epitope retrieval (HIER) using basic retrieval reagents has shown superior results compared to acidic solutions. For example, successful detection of IL-17/IL-17A in paraffin-embedded sections of human Crohn's disease intestine was achieved using heat-induced epitope retrieval with basic antigen retrieval reagent prior to antibody incubation .

For plant cell wall antibodies:

  • Enzymatic pretreatment with pectinases or hemicellulases may be necessary to expose masked epitopes in highly crosslinked plant cell wall structures

  • A sequential approach using sodium carbonate pretreatment followed by pectate lyase has shown enhanced detection of certain glycan epitopes in plant tissues

For optimal results, researchers should evaluate multiple retrieval methods in parallel, as effectiveness varies by:

  • Tissue type and fixation duration

  • Specific epitope and antibody clone

  • Target subcellular localization

Documentation of optimization experiments should include quantitative assessment of staining intensity and specificity for each method tested.

How can I troubleshoot non-specific binding when using 17 kDa cell wall antibodies in complex biological samples?

Non-specific binding represents a significant challenge when working with 17 kDa cell wall antibodies, particularly in heterogeneous samples. A systematic troubleshooting approach should include:

  • Antibody validation controls:

    • Include appropriate isotype controls (e.g., MAB002 for mouse monoclonals)

    • Perform peptide competition assays to confirm epitope specificity

    • Include positive and negative tissue controls in each experiment

  • Blocking optimization:

    • Test multiple blocking agents (BSA, serum, commercial blockers)

    • Extend blocking time to 2 hours at room temperature

    • Consider dual blocking with protein and detergent combinations

  • Washing optimization:

    • Increase washing duration and stringency

    • Test different detergent concentrations (0.1-0.5% Tween-20 or Triton X-100)

    • Consider high-salt washes (up to 500 mM NaCl) for electrostatic interactions

  • Secondary antibody cross-adsorption:

    • Use highly cross-adsorbed secondary antibodies

    • Test secondaries from multiple vendors

    • Consider directly conjugated primary antibodies to eliminate secondary cross-reactivity

For particularly challenging applications, such as detecting low-abundance 17 kDa proteins in complex microbial or plant samples, a signal amplification approach using tyramide signal amplification (TSA) may be beneficial while maintaining low primary antibody concentrations to minimize non-specific binding.

What advanced normalization strategies should be employed when quantifying Western blot data for 17 kDa cell wall proteins?

Quantitative analysis of 17 kDa cell wall proteins by Western blot requires robust normalization strategies to account for technical and biological variability. Based on research practices, the following advanced approaches are recommended:

  • Internal loading control selection:

    • Use housekeeping proteins with expression stability verified in your experimental system

    • α-tubulin and Hsp90 have been successfully used as internal controls for 17 kDa protein normalization

    • Consider multiple loading controls spanning different molecular weight ranges and subcellular compartments

  • Densitometric analysis parameters:

    • Utilize linear range calibration curves with purified standards

    • Perform pixel volume analysis rather than simple intensity measurements

    • Scan blots at 600 dpi using light transmittance for optimal resolution

  • Advanced normalization calculations:

    • Apply rolling ball background subtraction before quantification

    • Normalize target protein to multiple reference proteins using geometric averaging

    • Consider global normalization approaches for differentially expressed targets

  • Validation approaches:

    • Confirm key findings with orthogonal methods (ELISA, mass spectrometry)

    • Perform technical and biological replicates to establish variation coefficients

    • Use Bland-Altman plots to assess agreement between normalization methods

When analyzing membrane proteins or cell wall components, additional considerations include extraction efficiency normalization and detergent compatibility assessment to ensure representative sampling of the target proteins.

How should researchers design experiments to distinguish between specific 17 kDa protein isoforms with high sequence homology?

Distinguishing between closely related 17 kDa protein isoforms requires a multifaceted experimental approach that leverages both antibody specificity and complementary techniques:

  • Epitope mapping strategy:

    • Select antibodies targeting unique epitope regions not conserved between isoforms

    • Consider generating custom antibodies against synthetic peptides from divergent regions

    • Validate specificities using recombinant proteins expressing individual isoforms

  • Immunodepletion approach:

    • Sequential immunoprecipitation with isoform-specific antibodies

    • Quantitative analysis of depleted fractions

    • Confirmation by mass spectrometry of immunoprecipitated proteins

  • Expression system controls:

    • Use cell lines or tissues with known differential expression of specific isoforms

    • Include genetic knockouts or knockdowns of individual isoforms as validation controls

    • Consider heterologous expression systems for standardization

  • Advanced electrophoretic separation:

    • Employ high-resolution 2D gel electrophoresis to separate isoforms by both pI and molecular weight

    • Use extended gradient gels (e.g., 10-20%) for improved resolution in the 15-20 kDa range

    • Consider native gel conditions to leverage conformational differences between isoforms

For plant cell wall proteins, researchers should be aware that many antibodies recognize specific epitopes that may be present on multiple glycan classes. The comprehensive toolkit of plant cell wall glycan-directed mAbs has demonstrated that many antibodies recognize arabinose and/or galactose-containing structures present across different glycan classes . Therefore, these antibodies should be utilized as epitope-specific rather than polymer-specific probes.

What experimental controls are essential when validating a novel 17 kDa cell wall antibody for immunolocalization studies?

Rigorous validation of novel 17 kDa cell wall antibodies for immunolocalization requires a comprehensive set of controls to ensure specificity, sensitivity, and reproducibility:

  • Specificity controls:

    • Preincubation with immunizing peptide/protein (peptide competition)

    • Parallel staining with multiple antibodies against different epitopes of the same protein

    • Testing against samples with genetically modified expression (knockout/knockdown/overexpression)

    • Cross-reactivity assessment against related proteins in the same family

  • Technical controls:

    • Isotype controls matched to primary antibody class and concentration

    • Secondary antibody-only controls to assess non-specific binding

    • Endogenous peroxidase/phosphatase blocking validation for enzyme-based detection systems

    • Serial dilution series to establish optimal antibody concentration

  • Tissue processing controls:

    • Comparison of different fixation methods (aldehyde vs. alcohol-based)

    • Assessment of epitope retrieval methods (heat vs. enzymatic)

    • Positive control tissues with known expression patterns

    • Negative control tissues with confirmed absence of target

  • Complementary method validation:

    • Correlation with in situ hybridization for transcript localization

    • Comparison with fluorescent protein fusion localization patterns

    • Verification with subcellular fractionation and immunoblotting

    • Mass spectrometry confirmation of immunoprecipitated complexes

For plant cell wall antibodies specifically, hierarchical clustering analysis of binding patterns against diverse polysaccharide panels has been used to verify antibody specificity, with further validation through immunolocalization studies in model plants such as Arabidopsis stems .

What methodological considerations should be addressed when using 17 kDa cell wall antibodies across different species or tissue types?

Cross-species and cross-tissue applications of 17 kDa cell wall antibodies require careful methodological adaptations to account for biological and technical variables:

  • Epitope conservation assessment:

    • Perform sequence alignment analysis of target proteins across species

    • Focus on antibodies targeting highly conserved regions for cross-species work

    • Validate with recombinant proteins or synthetic peptides from each species

  • Tissue-specific extraction modifications:

    • Adjust extraction buffers based on tissue composition (lipid content, extracellular matrix density)

    • Optimize mechanical disruption methods for different tissue types

    • Consider tissue-specific protease inhibitor cocktails to prevent epitope degradation

  • Fixation and permeabilization adaptations:

    • Titrate fixative concentration and duration for each tissue type

    • Test graded permeabilization series for optimal antibody accessibility

    • Evaluate epitope stability under various fixation conditions

  • Signal detection optimization:

    • Adjust amplification systems based on target abundance in different tissues

    • Account for tissue-specific autofluorescence through appropriate controls and filters

    • Consider spectral imaging for tissues with complex autofluorescence profiles

When working with plant systems, researchers should recognize that transcript sizes for the same protein may vary across tissues. For example, Northern blot analysis revealed different transcript sizes in midgut (1.9 and 1.2 kb) versus Malpighian tubules (2.2 and 1.9 kb) for the same 17 kDa proteolipid . These tissue-specific variations must be accounted for in experimental design and interpretation.

How can researchers accurately quantify changes in 17 kDa cell wall protein expression across experimental conditions?

Accurate quantification of 17 kDa cell wall protein expression changes requires rigorous analytical approaches that account for various sources of technical and biological variability:

  • Sample preparation standardization:

    • Implement consistent extraction protocols with internal recovery standards

    • Normalize protein loading using total protein stains rather than single reference proteins

    • Account for extraction efficiency differences between experimental conditions

  • Quantitative detection strategies:

    • Employ fluorescent Western blotting for wider linear detection range

    • Utilize multiplex detection systems for simultaneous target and reference measurement

    • Establish standard curves with recombinant proteins for absolute quantification

  • Statistical analysis framework:

    • Apply appropriate normality tests before selecting parametric or non-parametric methods

    • Use ANOVA with post-hoc tests for multi-condition comparisons

    • Implement mixed-effects models to account for biological and technical replicates

  • Validation approaches:

    • Confirm protein-level changes with mRNA quantification

    • Utilize orthogonal quantification methods (ELISA, mass spectrometry)

    • Assess functional correlates of expression changes

For flow cytometry applications, researchers can quantify changes in 17 kDa protein expression by comparing median fluorescence intensity values between experimental conditions, as demonstrated in the analysis of IL-17 expression in human PBMCs after stimulation with PMA and Ca²⁺ ionomycin .

What methodological approaches can resolve contradictory results obtained using different antibodies targeting the same 17 kDa cell wall protein?

Resolving contradictory results from different antibodies targeting the same 17 kDa protein requires systematic investigation of both antibody characteristics and experimental variables:

  • Epitope mapping comparison:

    • Determine specific binding regions for each antibody using peptide arrays or deletion mutants

    • Assess whether epitopes may be differentially accessible in native conformations

    • Evaluate potential post-translational modification effects on epitope recognition

  • Methodological standardization:

    • Conduct parallel experiments with identical sample preparation, buffer systems, and detection methods

    • Test both antibodies against recombinant full-length protein and fragments

    • Evaluate performance across multiple application platforms (Western, IP, IHC)

  • Antibody validation assessment:

    • Review complete validation data for each antibody, including knockout/knockdown controls

    • Test for cross-reactivity against related proteins

    • Determine batch-to-batch variability through reference sample testing

  • Biological context analysis:

    • Investigate if discrepancies relate to specific cell types, tissues, or treatment conditions

    • Consider developmental or physiological states that might affect epitope accessibility

    • Evaluate potential protein-protein interactions that could mask specific epitopes

Resolution often requires complementary approaches, such as mass spectrometry or CRISPR-based genetic validation, to establish ground truth. For plant cell wall antibodies, discrepancies may arise because many mAbs recognize distinct epitopes present on the same glycan class, as observed with antibodies clustering in multiple subclades that bind to each glycan class .

How should researchers interpret differences in subcellular localization patterns detected by 17 kDa cell wall antibodies?

Interpreting subcellular localization differences detected by 17 kDa cell wall antibodies requires careful consideration of biological complexity and technical factors:

  • Resolution of biological mechanisms:

    • Evaluate developmental or cell cycle-dependent localization patterns

    • Consider stimulus-induced translocation (e.g., stress response, signaling activation)

    • Assess potential proteolytic processing that might separate domains with different localizations

  • Technical artifact elimination:

    • Compare multiple fixation and permeabilization protocols to rule out fixation artifacts

    • Utilize super-resolution microscopy techniques to resolve closely adjacent structures

    • Confirm with fractionation studies and organelle-specific markers

  • Antibody characteristic assessment:

    • Determine if antibodies recognize different post-translational modifications with distinct localizations

    • Evaluate if antibodies detect different isoforms or splice variants with unique targeting sequences

    • Consider epitope masking in specific subcellular compartments due to protein-protein interactions

  • Functional validation approaches:

    • Correlate localization patterns with known functional readouts

    • Use structure-function mutations to alter localization and assess antibody detection

    • Implement live-cell imaging with fluorescent fusion proteins to confirm dynamics

For plant cell wall components, apparent differences in localization may reflect the fact that many epitopes are present on multiple glycan classes with distinct distributions. Research has shown that epitopes recognized by certain mAbs, particularly those recognizing arabinose- and/or galactose-containing structures, are present on more than one glycan class .

What are the optimal protocols for using 17 kDa cell wall antibodies in multiplex immunofluorescence experiments?

Multiplex immunofluorescence with 17 kDa cell wall antibodies requires specialized protocols to achieve specific detection with minimal cross-reactivity. Based on empirical research, the following approach has proven most effective:

  • Antibody panel design considerations:

    • Select primary antibodies from different host species when possible

    • For same-species antibodies, use directly conjugated primaries with spectrally distinct fluorophores

    • Consider antibody isotypes and utilize isotype-specific secondary antibodies

    • Test each antibody individually before combining to verify specificity and optimal concentration

  • Sequential staining protocol:

    • Begin with the lowest abundance target using tyramide signal amplification (TSA)

    • Perform microwave treatment (95°C for 5 minutes in 10mM citrate buffer) between rounds to eliminate residual HRP activity

    • Progress from lowest to highest abundance targets

    • Include wash steps with high-salt buffer (500mM NaCl in PBS) between antibody incubations

  • Spectral overlap management:

    • Implement appropriate single-color controls for spectral unmixing

    • Use narrow bandpass filter sets to minimize bleed-through

    • Consider linear unmixing algorithms for closely overlapping fluorophores

    • Include autofluorescence controls for each tissue type

  • Validation controls:

    • Process serial sections with individual antibodies for comparison

    • Include fluorescence-minus-one (FMO) controls for each antibody in the panel

    • Perform replicate experiments with reversed staining order for key comparisons

Successful multiplex protocols have been implemented for detecting IL-17 alongside other immune markers in human tissues, enabling detailed spatial relationship analysis between IL-17-producing cells and their microenvironment .

What advanced techniques can be employed to improve the specificity and sensitivity of 17 kDa cell wall antibodies in challenging samples?

Improving sensitivity and specificity for 17 kDa cell wall antibodies in challenging samples requires advanced techniques that enhance signal while reducing background:

  • Signal amplification strategies:

    • Tyramide signal amplification (TSA) can increase sensitivity 10-100 fold

    • Polymer-HRP detection systems provide amplification with reduced background

    • Quantum dot conjugates offer photostable signal with minimal photobleaching

    • Proximity ligation assay (PLA) for detection of protein-protein interactions with single-molecule sensitivity

  • Background reduction approaches:

    • Implement dual blocking strategies (protein block followed by Fc-receptor block)

    • Pre-adsorb antibodies against tissue homogenates from negative control samples

    • Utilize automated microfluidic washing systems for improved stringency

    • Apply machine learning algorithms for automated background discrimination

  • Sample preparation enhancements:

    • Optimize fixation with dual crosslinkers (formaldehyde plus glutaraldehyde at low concentrations)

    • Implement pressure-assisted or ultrasound-enhanced antibody penetration

    • Utilize section thickness optimization based on target abundance (thinner for rare targets)

    • Consider tissue clearing techniques for deep tissue imaging

  • Antigen retrieval innovations:

    • Combine heat and enzymatic treatments in optimized sequences

    • Utilize high-pressure antigen retrieval systems for improved epitope exposure

    • Implement variable pH retrieval testing (pH 3-10) to identify optimal conditions

    • Consider ultrasonication-assisted epitope retrieval for resistant samples

For plant cell wall antibodies, hierarchical clustering analysis based on polysaccharide recognition patterns has been used to verify specificity across diverse plant materials, providing a powerful approach to identify the most specific mAbs for challenging applications .

How can researchers leverage 17 kDa cell wall antibodies for high-throughput screening applications in biomedical and agricultural research?

Adapting 17 kDa cell wall antibodies for high-throughput screening requires systematic optimization and standardization:

  • Assay miniaturization strategies:

    • Transition from standard ELISA to 384- or 1536-well microplate formats

    • Implement automated liquid handling systems for consistent antibody dispensing

    • Develop homogeneous assay formats to eliminate wash steps (e.g., TR-FRET, AlphaLISA)

    • Optimize reagent concentrations to maintain signal window while reducing costs

  • Detection platform considerations:

    • Utilize high-content imaging systems for subcellular resolution in cell-based screens

    • Implement multiplexed bead-based immunoassays for simultaneous detection of multiple targets

    • Develop label-free detection methods for real-time kinetic analyses

    • Consider microfluidic chip-based systems for reduced sample volume requirements

  • Quality control implementation:

    • Establish robust Z' factor metrics for each assay (aim for Z' > 0.5)

    • Incorporate internal calibration standards on each plate

    • Implement positive and negative controls in spatially distributed patterns

    • Develop standard operating procedures with decision trees for troubleshooting

  • Data analysis workflow development:

    • Create automated image analysis pipelines for consistent feature extraction

    • Implement machine learning algorithms for phenotypic classification

    • Develop dose-response curve fitting with appropriate statistical models

    • Establish database systems for storage, retrieval, and meta-analysis of screening results

Research has demonstrated the utility of plant cell wall glycan-directed mAbs in high-throughput glycome profiling, enabling rapid analysis of cell wall structure across diverse plant species and under various treatment conditions . Similar approaches can be adapted for biomedical applications, such as screening for modulators of IL-17 expression in human immune cells .

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.