34 kDa cell wall Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Significance

The term "34 kDa cell wall antibody" refers to immunoglobulins targeting proteins of approximately 34 kilodaltons (kDa) localized to microbial cell walls or surface-exposed antigens. These antibodies play critical roles in pathogen recognition, immune evasion, and therapeutic interventions across bacterial, fungal, and protozoan systems.

Bacterial Systems: Salmonella typhi

  • Antigen: A 34 kDa outer membrane protein (OMP) identified as highly antigenic in Salmonella typhi .

  • Epitope Localization: Surface-exposed regions recognized by patient-derived antibodies during typhoid fever .

  • Functional Role: Antibodies against this protein enhance opsonization and phagocytosis of intact bacteria .

Fungal Systems: Candida albicans

  • Target: Pga31, a 34 kDa glycosylphosphatidylinositol (GPI)-anchored cell wall protein critical for fungal adhesion and virulence .

  • Antibody Characteristics:

    • Monoclonal antibodies (mAbs) generated via phage display bind preferentially to hyphal forms .

    • Reformatted IgG mAbs show 400-fold higher affinity (EC₅₀ = 400 pM) compared to single-chain antibody fragments (scAbs) .

Protozoan Systems: Cryptosporidium parvum

  • Antigen: Cp-P34, a 34 kDa MORN (Membrane Occupation and Recognition Nexus) repeat protein transiently exposed on sporozoite surfaces .

  • Immune Response: Elicits antibodies in naturally infected alpacas, suggesting potential as a vaccine candidate .

Table 1: Comparative Analysis of 34 kDa Antibody Targets

OrganismAntigen NameLocalizationAntibody TypeKey Functional RoleReference
Salmonella typhi34 kDa OMPOuter membranePolyclonal (Patient sera)Opsonization, diagnostic marker
Candida albicansPga31Cell wall (hyphae)Monoclonal (1B11, 1H3)Enhances macrophage engulfment
Cryptosporidium parvumCp-P34Sporozoite surfaceVHH (Camelid-derived)Vaccine candidate, immune evasion

Table 2: Candida albicans Pga31 mAb Efficacy

Parameter1B11 scAb1B11 mAb
EC₅₀ (Peptide binding)175 nM400 pM
Hyphal binding affinityModerateHigh
Survival in murine modelN/A83% survival

Bacterial Systems

  • The 34 kDa OMP in Salmonella typhi is recognized by antibodies in 100% of typhoid patient sera, confirming its immunodominant nature .

  • Immunoblot assays demonstrate specificity for intact bacterial cells, suggesting surface accessibility .

Fungal Systems

  • Anti-Pga31 mAbs bind 3-fold more strongly to caspofungin-treated C. albicans hyphae, correlating with cell wall stress-induced epitope exposure .

  • Pre-treatment with mAbs reduces fungal burden by 2–3 logs in murine models of systemic candidiasis .

Protozoan Systems

  • Cp-P34 is shed in trails during sporozoite motility, facilitating immune evasion .

  • Antibodies against Cp-P34 block sporozoite attachment to host cells in vitro .

Implications for Therapeutics and Diagnostics

  • Vaccine Development: The 34 kDa OMP of Salmonella typhi and Cp-P34 of C. parvum are prioritized for subunit vaccine design .

  • Antifungal Therapy: Anti-Pga31 mAbs show synergistic effects with caspofungin, reducing therapeutic doses .

  • Diagnostic Tools: ELISA assays using purified 34 kDa antigens achieve >95% specificity in typhoid fever detection .

Challenges and Future Directions

  • Epitope Masking: Cell wall polysaccharides in C. albicans obscure Pga31 epitopes, necessitating enzymatic pre-treatment for antibody access .

  • Cross-Reactivity: VH4-34 antibodies in humans show autoreactivity to B cell surface glycans, complicating therapeutic use .

  • Species Specificity: The 34 kDa TA34 antigen in HTLV-I exhibits primate-specific epitopes, limiting broad applicability .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
34 kDa cell wall protein antibody; Fragment antibody
Uniprot No.

Target Background

Subcellular Location
Secreted, cell wall.

Q&A

What is the 34 kDa cell wall protein in Mycobacterium avium subsp. paratuberculosis?

The 34 kDa protein is a major species-specific immunodominant antigen found in the cell wall of Mycobacterium avium subsp. paratuberculosis (MAP), the causative agent of Johne's disease. This protein plays a significant role in the immune response against MAP infection and has been identified as a potential candidate for serodiagnostic tests . It is characterized by its strong immunogenicity properties, making it valuable for detecting MAP-specific antibodies in infected animals. The protein has been thoroughly studied for its antigenicity and has demonstrated promise as a biomarker for MAP infection due to its high specificity and sensitivity in diagnostic applications .

How does the 34 kDa cell wall protein compare to other MAP antigens in diagnostic applications?

Methodologically, the 34 kDa cell wall protein shows superior diagnostic performance compared to other MAP antigens. When evaluated against whole cell wall extracts and commercial diagnostic kits, the purified 34 kDa protein demonstrates exceptional specificity and sensitivity . In ELISA tests, serum samples from cattle confirmed to be infected with MAP consistently show positive results when tested with the 34 kDa antigen, while samples from healthy cattle consistently yield negative results . This clear differentiation between infected and uninfected animals highlights the protein's value in diagnostic research. To utilize this advantage, researchers should employ purified 34 kDa protein in ELISA-based formats with standardized protocols that include appropriate controls and validation steps to ensure reliable diagnostic outcomes.

What methods can be used to isolate the 34 kDa protein from bacterial cell walls?

The isolation of the 34 kDa protein from MAP cell walls involves a multi-step purification process. One effective method is electroelution, which has been successfully employed to isolate the cell wall-associated 34 kDa antigen . This technique involves separating the protein based on its molecular weight through electrophoresis followed by elution from the gel matrix. The process typically begins with the preparation of cell wall extracts from MAP cultures, followed by protein separation using SDS-PAGE. After identification of the 34 kDa band, electroelution is performed to recover the purified protein . This methodological approach ensures the isolation of the native protein in its biologically active form, which is crucial for subsequent immunological studies. Researchers should optimize buffer conditions and elution parameters to maximize protein recovery while maintaining structural integrity.

How does the immunogenicity of purified 34 kDa protein compare to whole cell wall extracts in experimental models?

The purified 34 kDa protein demonstrates superior immunogenicity compared to whole cell wall extracts in experimental models. In comparative ELISA tests, the 34 kDa antigen exhibits higher sensitivity and specificity than whole cell wall preparations . This enhanced performance is attributed to the elimination of cross-reactive epitopes that are present in complex antigen mixtures. Methodologically, researchers should implement a standardized ELISA protocol using the purified 34 kDa protein, including optimal antigen coating concentration (typically 1-5 μg/mL), appropriate blocking agents, and validated detection antibodies. When designing such experiments, it is essential to include positive and negative control sera from confirmed MAP-infected and healthy animals, respectively, along with commercially available reference standards for calibration . The experimental design should also incorporate statistical analysis to determine diagnostic cutoff values that maximize both sensitivity and specificity.

What are the structural and functional differences between the 34 kDa protein of MAP and its isolog in M. leprae?

While the 34 kDa proteins in MAP and M. leprae share isologous relationships, they exhibit distinct functional characteristics. The 34 kDa protein of MAP serves as an immunodominant antigen with strong serological activity, making it valuable for diagnostic applications . In contrast, the 34 kDa isolog in M. leprae, which also resides at the C terminus subcellular fractions, has been found to lack significant serological activity . This functional divergence despite structural similarity suggests evolutionary adaptations specific to the pathogenesis mechanisms of each mycobacterial species. From a methodological perspective, researchers investigating these differences should employ comparative genomic and proteomic approaches, including sequence alignment, epitope mapping, and three-dimensional structural analysis. Experimental designs should include recombinant protein expression systems to produce both proteins under identical conditions, followed by comparative immunological assays to quantify their respective antigenicity and reactivity with host immune components.

How can epitope mapping of the 34 kDa protein enhance antibody development strategies?

Epitope mapping of the 34 kDa protein can significantly enhance antibody development by identifying the specific antigenic determinants that elicit strong immune responses. Previous studies have demonstrated that the 34 kDa protein contains multiple B-cell epitopes recognized by antibodies from infected cattle . Methodologically, researchers can employ various approaches for epitope mapping, including peptide microarray analysis, phage display libraries, and computational prediction algorithms combined with experimental validation. The identified epitopes can then be synthesized as peptides and used to develop highly specific monoclonal antibodies through hybridoma technology or phage display systems . This targeted approach allows for the development of antibodies with enhanced specificity and reduced cross-reactivity with related mycobacterial species. Furthermore, identified epitopes can be incorporated into multi-epitope vaccine candidates or diagnostic platforms with improved performance characteristics.

What are the optimal conditions for using the 34 kDa protein in serodiagnostic ELISA tests?

The optimal conditions for using the 34 kDa protein in serodiagnostic ELISA tests involve several critical parameters that must be carefully controlled. The purified protein should be coated onto high-binding ELISA plates at a concentration of 1-5 μg/mL in carbonate-bicarbonate buffer (pH 9.6) overnight at 4°C . Blocking should be performed with 1-3% bovine serum albumin or non-fat dry milk in PBS-T (PBS containing 0.05% Tween-20) to minimize non-specific binding. Serum samples should be diluted appropriately (typically 1:100 to 1:500) in blocking buffer and incubated for 1-2 hours at room temperature or 37°C . After washing, species-appropriate enzyme-conjugated secondary antibodies should be applied, followed by colorimetric substrate development and absorbance measurement at the appropriate wavelength. Methodologically, researchers should perform optimization experiments to determine the ideal antigen concentration, serum dilution, and incubation times that maximize the signal-to-noise ratio and diagnostic accuracy for their specific application.

How can researchers address cross-reactivity issues when developing antibodies against the 34 kDa protein?

Addressing cross-reactivity issues requires a multi-faceted methodological approach. Researchers should first perform comprehensive sequence analysis to identify regions unique to the MAP 34 kDa protein that are absent or significantly different in related mycobacterial species . These unique regions can then be synthesized as peptides and used for immunization or antibody selection. Affinity purification techniques using immobilized MAP-specific peptides can be employed to isolate antibodies that recognize only the target epitopes. Additionally, extensive cross-adsorption against antigens from related mycobacterial species can eliminate antibodies with cross-reactive potential . From an experimental design perspective, researchers should include a panel of controls representing related mycobacterial species and environmental mycobacteria to validate antibody specificity. Implementing these methodological strategies can significantly reduce cross-reactivity while maintaining high sensitivity for the target 34 kDa protein.

What are the molecular determinants of the high specificity of the 34 kDa protein as a diagnostic marker?

The high specificity of the 34 kDa protein as a diagnostic marker is attributed to several molecular determinants. Studies have shown that this protein contains unique sequence regions and conformational epitopes that are specific to MAP and not shared with other mycobacterial species . Additionally, post-translational modifications specific to MAP may contribute to the protein's antigenicity profile. Methodologically, researchers investigating these determinants should employ a combination of approaches including computational analysis of protein sequence and structure, experimental characterization of protein modifications, and systematic epitope mapping using both linear peptides and conformational mimetics . Recombinant protein expression systems can be used to produce variants with specific mutations or truncations to identify the critical regions for antigenicity and specificity. Mass spectrometry analysis can reveal post-translational modifications that may contribute to the protein's diagnostic utility. Understanding these molecular determinants can guide the rational design of improved diagnostic reagents with enhanced specificity and sensitivity.

How should researchers design validation studies for new 34 kDa protein-based diagnostic tests?

Validation studies for 34 kDa protein-based diagnostic tests require rigorous experimental design and statistical analysis. Researchers should use a panel of well-characterized samples from confirmed MAP-infected animals (based on culture and/or PCR) and definitively negative controls from MAP-free herds . The sample size should be determined through power analysis to ensure statistical significance, typically including at least 50-100 samples in each group representing various stages of infection. The experimental design should include comparison with gold standard methods and established commercial tests. Methodologically, researchers should implement a blinded testing protocol where sample identities are unknown to the test performers to eliminate bias. Statistical analysis should include calculation of sensitivity, specificity, positive and negative predictive values, and receiver operating characteristic (ROC) curve analysis to determine optimal cutoff values . Additionally, inter- and intra-assay variability should be assessed to determine reproducibility. This comprehensive validation approach ensures that new diagnostic tests based on the 34 kDa protein meet the rigorous standards required for research and potential clinical applications.

What methodological approaches can be used to improve the solubility and stability of the 34 kDa protein for immunoassay development?

Improving the solubility and stability of the 34 kDa protein requires specific methodological strategies due to its cell wall association. Researchers can employ detergent-based extraction methods using mild non-ionic detergents like Triton X-100 or octyl glucoside to solubilize the protein while maintaining its native conformation . Alternative approaches include the use of recombinant protein expression systems with solubility-enhancing fusion partners such as maltose-binding protein (MBP) or thioredoxin. For long-term stability, the purified protein should be stored in buffer systems containing stabilizing agents such as glycerol (10-20%), trehalose, or bovine serum albumin. Lyophilization following addition of appropriate cryoprotectants represents another viable approach for long-term storage. To monitor protein stability, researchers should implement regular quality control testing including SDS-PAGE, Western blotting, and functional assays to assess immunoreactivity . The development of a standardized production and storage protocol is essential for ensuring consistent performance in immunoassay applications and facilitating comparison of results across different studies.

How can discrepancies in serological results between different 34 kDa protein-based assays be explained and resolved?

Discrepancies in serological results between different 34 kDa protein-based assays can arise from multiple sources including variations in protein preparation, assay formats, and analytical parameters. To explain and resolve these discrepancies, researchers should first conduct a systematic comparison of the protein preparations used in different assays, examining purity, conformation, and epitope presentation using techniques such as SDS-PAGE, Western blotting, and epitope mapping . Next, assay protocols should be standardized with respect to critical parameters including antigen coating concentration, blocking conditions, sample dilution, incubation times and temperatures, and detection systems. A panel of reference samples with established serological status should be tested across all assay platforms to calibrate results . Methodologically, researchers should implement statistical approaches such as Bland-Altman analysis to quantify agreement between methods and identify systematic biases. When possible, collaborative studies involving multiple laboratories should be conducted to assess interlaboratory reproducibility. By systematically addressing these factors, researchers can identify the sources of discrepancies and develop harmonized protocols that yield consistent results across different research settings.

What are the potential applications of monoclonal antibodies against the 34 kDa protein beyond diagnostic tests?

Monoclonal antibodies against the 34 kDa protein have potential applications extending well beyond diagnostic tests. From a methodological perspective, these antibodies can be used to develop immunoaffinity purification systems for isolating the native protein from complex mycobacterial extracts, facilitating detailed structural and functional studies . They can also be employed in immunohistochemistry and immunofluorescence techniques to study the localization and expression patterns of the protein in infected tissues, providing insights into pathogenesis mechanisms. In research settings, these antibodies can be used to develop immunoprecipitation protocols for identifying interaction partners of the 34 kDa protein, potentially revealing its role in bacterial physiology and host-pathogen interactions . Additionally, they may serve as valuable tools for monitoring MAP environmental contamination through the development of sensitive detection methods for food and water samples. From a therapeutic perspective, if the 34 kDa protein is found to play a critical role in pathogenesis, antibodies targeting this protein could potentially be developed into passive immunotherapy approaches or used to guide vaccine design strategies.

How might comparative proteomic approaches enhance our understanding of the 34 kDa protein's role in pathogenesis?

Comparative proteomic approaches can significantly enhance our understanding of the 34 kDa protein's role in pathogenesis by revealing its expression patterns, post-translational modifications, and interaction networks across different conditions and mycobacterial species. Methodologically, researchers can employ quantitative proteomics techniques such as iTRAQ, SILAC, or label-free quantification to compare protein abundance in virulent versus attenuated MAP strains, or in bacteria grown under conditions mimicking different stages of infection . Protein interaction studies using approaches such as pull-down assays, co-immunoprecipitation followed by mass spectrometry, or bacterial two-hybrid systems can identify binding partners of the 34 kDa protein, potentially revealing its functional networks. Structural proteomics techniques including X-ray crystallography or cryo-electron microscopy can elucidate the three-dimensional structure of the protein, providing insights into its potential functional domains. Additionally, post-translational modification analysis can reveal how modifications such as glycosylation or phosphorylation might regulate the protein's function or immunogenicity. These comprehensive proteomic approaches can establish connections between the 34 kDa protein and specific pathogenesis mechanisms, potentially identifying new targets for therapeutic intervention.

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.