ALA9 Antibody

Shipped with Ice Packs
In Stock

Description

1.1. Potential Typographical Errors

  • AAV9 Antibody: A well-documented antibody target in gene therapy (e.g., adeno-associated virus serotype 9).

    • Relevance: Anti-AAV9 antibodies are critical in gene therapy for diseases like spinal muscular atrophy (SMA). Elevated AAV9 antibody titers can exclude patients from treatment with onasemnogene abeparvovec .

    • Key Data:

      ParameterValueSource
      Seroprevalence in pediatric SMA patients13.04% (elevated first test)
      Epitope regions on AAV9 capsid3-fold, 2/5-fold, 5-fold axes

1.2. Antilymphocyte Antibody (ALA)

  • Context: Antilymphocyte antibodies (ALAs) are associated with immune deficiencies.

    • Findings: High ALA levels correlate with progression to AIDS in HIV-positive patients .

1.3. Hypothetical Construct

  • If referring to a novel antibody, no structural, functional, or clinical data exist in indexed literature.

2.1. AAV9-Targeting Antibodies

  • Function: Neutralize AAV9 vectors used in gene therapy.

  • Structural Insights:

    • Epitopes mapped to capsid regions near 3-fold/5-fold axes .

    • Critical residues: S454, P659 (confirmed via cryo-EM and mutagenesis) .

  • Clinical Impact:

    AAV9 VariantNeutralization EscapeTransduction Efficiency
    S454A/P659AYes (vs. ADK9, HL2370)Maintained/Improved

2.2. Anti-HLA-E Antibodies

  • Example: mAb 3H4v31 enhances NK cell tumor killing via ADCC .

    • Affinity: 700-fold improvement over parent antibody .

3.1. Antibody Engineering Databases

  • AB-Bind Database: Contains 1,101 mutants with ΔΔG values for antibody-antigen interactions .

  • Therapeutic Antibodies: 162 FDA-approved products as of 2024, none named "ALA9" .

3.2. Antibody Structure-Function Relationships

  • Key Domains:

    • Fab: Binds antigen via CDR loops (e.g., CDR-H3 contributes 40–60% of binding energy) .

    • Fc: Mediates effector functions (e.g., ADCC via FcγRIIIa) .

Recommendations for Further Inquiry

  1. Verify Terminology: Confirm whether "ALA9" refers to AAV9, ALA, or a proprietary compound not yet published.

  2. Explore Patent Databases: Unpublished antibodies may appear in patent filings (e.g., WO/2024/123456).

  3. Clinical Trials: Search ClinicalTrials.gov for ongoing studies using advanced filters (e.g., "monoclonal antibody" + "ALA9").

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
ALA9 antibody; At1g68710 antibody; F24J5.6 antibody; Putative phospholipid-transporting ATPase 9 antibody; AtALA9 antibody; EC 7.6.2.1 antibody; Aminophospholipid flippase 9 antibody
Target Names
ALA9
Uniprot No.

Target Background

Function
ALA9 Antibody is involved in the transport of phospholipids.
Database Links

KEGG: ath:AT1G68710

STRING: 3702.AT1G68710.1

UniGene: At.52438

Protein Families
Cation transport ATPase (P-type) (TC 3.A.3) family, Type IV subfamily
Subcellular Location
Membrane; Multi-pass membrane protein.

Q&A

What are natural IgM Anti-Leukocyte Auto-Antibodies and their primary functions?

Natural IgM Anti-Leukocyte Auto-Antibodies (IgM-ALA) are a heterogeneous group of several different antibodies that react with various receptors present on autologous and allogeneic leukocytes and other cells expressing leukocyte receptors. These naturally occurring antibodies are encoded by minimally or non-mutated germline genes, making them characteristically polyreactive with low binding affinity. A critical distinguishing feature of these IgM-ALA is that they do not mediate cytolysis in the presence of complement at body temperature, differentiating them from disease-producing auto-antibodies which are predominantly IgG isotype with high affinity and specificity. Studies with human B cell clones from umbilical cord demonstrate that only approximately 10 percent of IgM secreting clones exhibit IgM-ALA reactivity, and these clones display different receptor specificities. Research has shown that IgM isolated from human serum can immunoprecipitate CD3 and CD4, inhibit T cell activation/proliferation, and suppress leukocyte production of certain cytokines such as TNF-α .

How do antibodies like IgM-ALA interact with cellular receptors?

IgM-ALA bind to various undefined membrane receptors comprising glycoproteins, phospholipids, and glycolipids. Their interaction mechanisms include immunoprecipitation of chemokine receptors such as CXCR4 and CCR5, inhibition of the binding of chemokines to these receptors, and subsequent inhibition of chemotaxis induced by chemokines. In in-vitro studies, purified murine serum IgM (but not IgM pre-absorbed with splenic leukocytes to remove IgM-ALA) has been shown to inhibit the production and activation of pro-inflammatory cells involved in both innate and adaptive immune responses. Additionally, these antibodies inhibit TLR-4, which plays a crucial role in initiating inflammatory processes. This inhibitory action on TLR-4 receptors occurs specifically on antigen-presenting cells (APC) and endothelial cells, demonstrating the specialized cellular targets of these antibodies .

What are the key differences between natural antibodies and disease-producing auto-antibodies?

Natural antibodies such as IgM-ALA differ fundamentally from disease-producing auto-antibodies in several important aspects. Natural antibodies are predominantly of the IgM isotype, encoded by minimally mutated germline genes, display polyreactivity with low binding affinity, and crucially, do not mediate cytolysis at body temperature (37°C). In contrast, disease-producing auto-antibodies are primarily of the IgG isotype, bind with high affinity and specificity to their auto-antigens, and can mediate cytolysis at physiological temperatures. This distinction is critical when designing experiments to investigate the protective versus pathological roles of different antibody classes. Additionally, natural antibodies often serve regulatory functions in the immune system, inhibiting T cell activation and cytokine production, whereas pathological auto-antibodies typically promote inflammation and tissue damage through specific targeting of self-antigens .

How can researchers validate antibody specificity and performance consistency across experiments?

Antibody validation represents one of the most challenging aspects of antibody-based research, contributing significantly to the reproducibility crisis in biomedical science. A robust validation approach requires distinguishing between testing data and validation data, with particular attention to consistency between batches and aliquots. Researchers should implement a two-tier approach that enables anticipation of how an antibody will likely perform when repeated purchases are required. For specificity validation, techniques should include western blotting with appropriate controls, immunoprecipitation followed by mass spectrometry, testing in knockout/knockdown systems, and cross-validation with multiple antibodies targeting different epitopes of the same protein. Performance consistency can be ensured through systematic documentation of lot-to-lot variation, regular antibody titration, inclusion of standardized positive and negative controls in each experiment, and maintenance of detailed records of storage conditions and freeze-thaw cycles that might affect antibody performance .

What methodological approaches best characterize pre-existing and treatment-emergent anti-antibody responses?

Characterization of pre-existing and treatment-emergent anti-antibody responses requires sensitive and drug-tolerant assay systems. A particularly effective approach is the development of affinity capture and elution (ACE) assays with labeled detection systems. For example, in the context of adeno-associated virus (AAV) based gene therapies, researchers have developed a total antibody (TAb) assay using recombinant AAV9-GFP as a surrogate, with ruthenium-labeled AAV9-GFP for detection. This method achieved a sensitivity of 11.2 ng/mL and demonstrated excellent drug tolerance (5.4 × 10^10 DRP/mL AAV9-GFP at 100 ng/mL anti-AAV9 antibodies). When developing similar assays for other antibody systems, researchers should evaluate multiple commercial monoclonal antibodies as potential positive control materials, assess assay specificity through cross-reactivity studies, and validate precision through intra- and inter-assay coefficient of variation determinations. Additionally, screening normal human sera can provide valuable baseline data for identifying significant pre-existing antibodies that might interfere with therapeutic applications .

How do antibodies like IgM-ALA influence inflammatory processes in various disease models?

IgM-ALA play a crucial regulatory role in inflammatory processes across various disease models. In murine models of acute inflammation, such as heart allograft rejection (mediated by allopeptide-activated T cells) and kidney ischemia reperfusion injury (mediated by NK and NKT cells), IgM-ALA demonstrate significant anti-inflammatory effects. Mechanistically, these antibodies inhibit TLR-4, a receptor that initiates inflammatory cascades when activated by endogenous ligands released after tissue damage. Research has shown that purified murine serum IgM containing IgM-ALA, but not IgM pre-absorbed with splenic leukocytes to remove IgM-ALA, inhibits the production and activation of pro-inflammatory cells involved in both innate and adaptive immune responses. Tissue section analyses using specific antibodies (including anti-neutrophil, anti-Foxp3, anti-IL17A, anti-CD31, anti-F4/80, anti-TLR-4/MD-2, anti-murine JE, and anti-murine KC) have revealed the cellular targets of IgM-ALA and their effects on inflammatory mediator expression. This research highlights the potential therapeutic applications of natural antibodies in controlling inflammatory conditions .

What factors should researchers consider when designing antibody-based experiments?

When designing antibody-based experiments, researchers must carefully consider several critical factors to ensure reliable and reproducible results. First, antibody selection should be based on comprehensive validation data rather than vendor claims alone, with preference given to antibodies validated specifically for the intended application (e.g., Western blot, immunoprecipitation, or immunohistochemistry). Second, researchers should implement appropriate controls, including isotype controls, blocking peptides, and knockout/knockdown samples when available. Third, experimental conditions must be optimized for each specific antibody, including concentration, incubation time, temperature, and buffer composition. Fourth, researchers should document and control for batch-to-batch variability by maintaining detailed records of lot numbers and performing validation tests on new lots. Finally, researchers must consider potential cross-reactivity with unintended targets, particularly when working with polyclonal antibodies or in complex biological samples with multiple related proteins .

How can researchers optimize antibody performance for specific applications?

Optimizing antibody performance requires systematic approach tailored to the specific application. For immunohistochemistry applications, researchers should test multiple fixation methods (formalin, methanol, acetone) and antigen retrieval techniques (heat-induced versus enzymatic) to determine optimal conditions for each antibody. When using antibodies for flow cytometry, titration experiments are essential to identify the concentration that provides maximum signal-to-noise ratio, while blocking steps should be optimized to minimize non-specific binding. For Western blotting applications, researchers should test various blocking agents (BSA, milk, commercial blockers) as certain antibodies perform poorly with specific blockers. In immunoprecipitation experiments, optimization of bead type, antibody-to-bead ratio, and washing stringency is crucial for success. Additionally, researchers should consider whether direct labeling of antibodies would improve performance by eliminating secondary antibody cross-reactivity issues. These optimization steps should be systematically documented and standardized within research groups to ensure consistent performance across experiments .

What specialized detection systems enhance antibody sensitivity in complex biological samples?

Advanced detection systems can significantly enhance antibody sensitivity in complex biological samples. Affinity capture and elution (ACE) systems with labeled detection reagents represent a particularly effective approach, as demonstrated in the detection of anti-AAV9 antibodies using ruthenium-labeled AAV9-GFP. This method achieved an impressive sensitivity of 11.2 ng/mL while maintaining excellent drug tolerance. For immunohistochemistry applications, tyramide signal amplification (TSA) systems can enhance detection sensitivity up to 100-fold compared to conventional methods, enabling visualization of low-abundance targets. In flow cytometry, quantum dots and polymer-based detection systems offer superior brightness and photostability compared to traditional fluorophores. For Western blotting, chemiluminescent substrates with enhanced sensitivity or near-infrared fluorescent detection systems provide superior quantitative range and sensitivity. Additionally, digital PCR-coupled proximity ligation assays represent an emerging technology that can detect antibody binding with femtomolar sensitivity. Researchers should select detection systems based on both the abundance of the target antigen and the specific requirements of their experimental system .

How should researchers address batch-to-batch variability in antibody performance?

Batch-to-batch variability represents a significant challenge in antibody research that directly impacts reproducibility. Researchers should implement a comprehensive strategy to address this issue. First, establish a standardized validation protocol for each new antibody lot, including specificity testing with appropriate positive and negative controls. Second, maintain a reference stock of well-characterized antibody lots for direct comparison with new batches. Third, when planning long-term studies, consider purchasing sufficient quantities of a single lot or requesting reservation of specific lots from vendors. Fourth, implement internal reference standards and normalization procedures in all experiments to account for potential variations in antibody performance. Fifth, document all lot numbers, storage conditions, and handling procedures in laboratory notebooks and publications. Finally, consider developing a pooled antibody approach for critical experiments, where multiple batches are combined to average out batch-specific variations. These practices should be formalized within research groups to create a systematic approach to managing antibody variability .

What statistical approaches are most appropriate for analyzing antibody binding and affinity data?

Statistical analysis of antibody binding and affinity data requires specialized approaches to account for the unique characteristics of antibody-antigen interactions. For equilibrium binding studies, non-linear regression models using the appropriate binding equation (one-site, two-site, or cooperative binding) should be employed rather than linear transformations that can distort error structures. When analyzing antibody mutations and their effects on binding affinity, correlation metrics such as Pearson (r) and Spearman (ρ) coefficients provide valuable insights, as demonstrated in the DyAb antibody design system which achieved correlation coefficients of r = 0.84 and ρ = 0.84 for anti-IL-6 variant test sets. For comparing multiple antibodies or conditions, ANOVA with appropriate post-hoc tests (Tukey's or Dunnett's) should be used rather than multiple t-tests to control family-wise error rates. When working with small sample sizes, non-parametric tests such as Mann-Whitney U or Kruskal-Wallis provide more robust analysis than parametric alternatives. Additionally, researchers should report effect sizes alongside p-values to provide a complete picture of the biological significance of their findings .

How can researchers effectively differentiate between specific and non-specific antibody interactions?

Differentiating between specific and non-specific antibody interactions requires a multi-faceted approach combining experimental controls and analytical methods. Competitive binding assays represent a powerful technique, where excess unlabeled antigen is used to compete with labeled antigen; specific interactions will show dose-dependent reduction in binding while non-specific interactions will not. Sequential immunoprecipitation provides another robust approach, where samples are first depleted with the antibody of interest, followed by a second immunoprecipitation - absence of signal in the second precipitation supports specificity. Knockout or knockdown validation is the gold standard, where antibody reactivity should be absent or significantly reduced in samples lacking the target protein. Cross-validation with multiple antibodies targeting different epitopes of the same protein can provide strong evidence of specificity when they show concordant results. Statistical approaches such as comparing signal-to-noise ratios across different conditions can help quantify specificity, with specific interactions typically showing higher ratios than non-specific ones. Finally, mass spectrometry analysis of immunoprecipitated proteins can provide unbiased identification of all proteins bound by an antibody, revealing potential cross-reactivity .

How might artificial intelligence transform antibody design and characterization?

Artificial intelligence is poised to revolutionize antibody design and characterization through several innovative approaches. Recent developments, such as the DyAb model, demonstrate the potential of AI in predicting antibody properties based on sequence information, achieving impressive correlation coefficients (r = 0.84, ρ = 0.84) for affinity predictions of antibody variants. Beyond prediction, AI systems can generate novel antibody variants with enhanced properties; in one study, 85% of AI-designed antibodies successfully expressed and bound to target antigens, with 84% showing improved affinity compared to the parent antibody. Vanderbilt University Medical Center's ambitious project, supported by up to $30 million from ARPA-H, aims to build a massive antibody-antigen atlas and develop AI-based algorithms to engineer antigen-specific antibodies, potentially addressing significant bottlenecks in traditional antibody discovery processes. These AI approaches could democratize antibody development, allowing researchers to efficiently generate monoclonal antibody therapeutics against targets of interest without the limitations of traditional discovery methods, which suffer from inefficiency, high costs, logistical hurdles, long turnaround times, and limited scalability .

What role might natural antibodies like IgM-ALA play in developing novel therapeutic approaches?

Natural antibodies such as IgM-ALA show considerable promise for developing novel therapeutic approaches based on their unique immunomodulatory properties. These antibodies demonstrate significant anti-inflammatory effects by inhibiting TLR-4 signaling on antigen-presenting cells and endothelial cells, thereby suppressing the initiation of inflammatory cascades. In murine models of acute inflammation, including heart allograft rejection and kidney ischemia reperfusion injury, IgM-ALA effectively attenuated inflammatory responses by inhibiting the production and activation of pro-inflammatory cells involved in both innate and adaptive immune responses. These properties suggest potential therapeutic applications in transplantation medicine, autoimmune disorders, and ischemia-reperfusion injuries. Furthermore, the natural occurrence of these antibodies might reduce immunogenicity concerns associated with engineered therapeutic antibodies. Future therapeutic development could focus on isolating specific IgM-ALA subtypes with enhanced anti-inflammatory properties, engineering synthetic versions with optimized pharmacokinetic profiles, or developing strategies to stimulate endogenous production of these protective antibodies in patients with inflammatory conditions .

How will developments in antibody validation standards impact biomedical research reproducibility?

Improvements in antibody validation standards will significantly impact biomedical research reproducibility by addressing one of the root causes of the current reproducibility crisis. The development of comprehensive validation frameworks that distinguish between testing data and validation data will provide researchers with more reliable information about antibody performance. Industry-wide adoption of standardized reporting formats for antibody characteristics would enable researchers to make more informed decisions when selecting reagents for their experiments. The implementation of two-tier approaches for antibody validation, which enable scientists to anticipate how an antibody will perform in repeated purchases, will reduce variability in experimental results. Additionally, the establishment of independent validation repositories and resources, where antibody performance is systematically documented across different applications and conditions, will provide the research community with unbiased information about reagent reliability. Collectively, these developments in validation standards will reduce experimental variability, enhance data quality, facilitate cross-laboratory comparisons, and ultimately accelerate scientific progress by ensuring that findings based on antibody-dependent methods are both robust and reproducible .

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.