DIR11 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
DIR11 antibody; At1g22900 antibody; F19G10.14Dirigent protein 11 antibody; AtDIR11 antibody
Target Names
DIR11
Uniprot No.

Target Background

Function
DIR11 proteins are key enzymes in plant secondary metabolism. They exhibit stereoselectivity in the phenoxy radical-coupling reaction, enabling the formation of optically active lignans. This reaction involves the coupling of two coniferyl alcohol molecules, leading to the biosynthesis of various compounds, including lignans, flavonolignans, and alkaloids.
Database Links

KEGG: ath:AT1G22900

STRING: 3702.AT1G22900.1

UniGene: At.41578

Protein Families
Plant dirigent protein family
Subcellular Location
Secreted, extracellular space, apoplast.

Q&A

What is CD11d and why is it a valuable therapeutic target?

CD11d is an alpha chain that forms part of the β2 integrin CD11d/CD18, which is expressed on leukocytes and plays a critical role in inflammatory cell migration. This integrin is a valuable therapeutic target because it mediates leukocyte extravasation in inflammatory conditions, including neurotrauma, sepsis, and atherosclerosis. Targeting CD11d/CD18 using monoclonal antibodies has been shown to reduce inflammation by modulating leukocyte migration into affected tissues, particularly in cases of acute neurotrauma .

How are humanized anti-CD11d monoclonal antibodies developed?

Humanized anti-CD11d monoclonal antibodies are developed through a process that combines the complementarity-determining regions (CDRs) of mouse-derived antibodies with human antibody frameworks. The process typically begins with the creation of murine antibodies that target the ligand-binding α-I domain of human CD11d. These murine antibodies are then humanized by transferring their CDRs to a human IgG framework (commonly IgG4), resulting in antibodies with reduced immunogenicity while maintaining target specificity .

How can researchers verify the specificity of CD11d antibodies?

Researchers can verify antibody specificity through multiple complementary approaches:

  • Flow cytometry analysis using cell types known to express CD11d (monocytes, neutrophils) and negative control cells (such as Jurkat T cells that do not express CD11d)

  • Competition assays with known ligands or other antibodies with established binding sites

  • Western blotting to confirm binding to the target protein at the expected molecular weight

  • Immunohistochemistry on tissues with known CD11d expression patterns

  • Comparing binding patterns to previously validated antibodies against the same target

What cell types express CD11d and how does expression vary between cell populations?

CD11d is primarily expressed on leukocytes, with varying levels of expression across different cell subsets. According to flow cytometry analysis, nonclassical CD14+CD16+ monocytes exhibit the highest level of surface-expressed CD11d among monocyte subsets. Both monocytes and neutrophils express CD11d, while T cells positive for the αβ T-cell receptor do not express CD11d. Understanding these expression patterns is crucial for predicting therapeutic efficacy and designing appropriate experimental controls .

How do conformational states of CD11d/CD18 affect antibody binding and function?

CD11d/CD18 integrins exist in active and inactive conformational states, which can dramatically affect antibody binding and function. Some humanized anti-CD11d antibodies (like anti-CD11d-2 clone) bind CD11d regardless of its conformational state, as demonstrated by binding studies in the presence of Mn²⁺ (which promotes the active conformation) or EDTA (which promotes the inactive conformation). This promiscuous conformational binding allows these antibodies to target both inactive CD11d/CD18 on peripheral blood leukocytes and active CD11d/CD18 on tissue-recruited leukocytes, potentially offering broader therapeutic coverage .

The binding epitope location plays a crucial role in this behavior. For instance, antibodies targeting regions near the α7-helix (which elongates upon divalent cation binding) may be more sensitive to conformational changes than those binding to other regions of the integrin .

What mechanisms underlie the therapeutic efficacy of anti-CD11d antibodies in neurotrauma models?

The therapeutic efficacy of anti-CD11d antibodies in neurotrauma models involves multiple mechanisms:

  • Inhibition of leukocyte extravasation: By targeting CD11d/CD18, these antibodies reduce the migration of inflammatory leukocytes into injured neural tissues.

  • Competition with natural ligands: Anti-CD11d antibodies that compete with ACE2 or other natural ligands can block signaling pathways that promote inflammation.

  • Modulation of integrin-mediated signaling: Some antibodies bind CD11d without inducing outside-in signaling, preventing inflammatory cascade activation.

  • Potential antibody-dependent cellular cytotoxicity (ADCC): Certain antibody isotypes (like IgG1) may mediate moderate ADCC, contributing to the elimination of inflammatory cells .

In rat spinal cord injury models, anti-CD11d treatment has demonstrated significant improvement in neurological and behavioral recovery, likely through these combined mechanisms .

How can researchers differentiate between total and surface-level expression of CD11d?

Research using anti-CD11d-2 as a detection tool has uncovered a mismatch between total and surface-level CD11d and CD18 expression. This distinction is critical for understanding the functional availability of the integrin. Researchers can differentiate between these expression levels through:

  • Surface expression: Flow cytometry on non-permeabilized cells detects only surface-expressed protein

  • Total expression: Western blotting of whole cell lysates or flow cytometry with permeabilized cells reveals the total protein pool

  • Subcellular fractionation: Separation of membrane and cytoplasmic fractions followed by immunoblotting

  • Imaging techniques: Confocal microscopy with and without permeabilization to visualize different protein populations

The regulation of surface expression (versus total cellular content) appears to be complex and not simply controlled by CK2 inhibition, suggesting multiple regulatory mechanisms control CD11d display on cell surfaces .

What are the optimal methods for evaluating anti-CD11d antibody binding characteristics?

MethodApplicationKey ParametersAdvantages
Flow CytometryCell binding assaysPercentage of positive cells; Mean Fluorescence Intensity (MFI)Quantifies binding to native receptors on live cells
BLI/SPRBinding kineticsAssociation/dissociation rates; KD valuesProvides real-time binding data without labels
Competition AssaysEpitope mappingIC50 values; Percent inhibitionDetermines relationship to known binding sites
Conformational StudiesBinding mechanismBinding in presence of Mn²⁺ or EDTAReveals conformation-dependent binding
Western BlottingSpecificity verificationMolecular weight; Band intensityConfirms target identity and expression levels

For comprehensive characterization, researchers should employ multiple complementary methods. For example, flow cytometry demonstrated that the humanized anti-CD11d-2 clone bound human monocytes and neutrophils with the greatest percentage and MFI among tested clones .

What animal models are suitable for testing the efficacy of CD11d-targeting antibodies?

Several animal models have proven valuable for testing anti-CD11d antibodies:

  • Rat spinal cord injury (SCI) model: This model has successfully demonstrated the therapeutic benefits of both murine and humanized anti-CD11d antibodies. SCI is induced through standardized compression injury, followed by antibody administration and assessment of neurological recovery using validated locomotor scoring systems .

  • Mouse models of SARS-CoV-2 infection: While not specific to CD11d studies, these models demonstrate principles applicable to antibody therapeutic evaluation:

    • Mouse ACE2-adapted virus models in wild-type BALB/c mice

    • Transgenic mice expressing human ACE2

    • Hamster models for both prophylactic and therapeutic efficacy assessment

The choice of model should align with the specific pathology being targeted. For neurotrauma applications, rat SCI models have consistently demonstrated that anti-CD11d antibodies improve neurological outcomes .

How can researchers optimize humanization strategies for therapeutic antibodies?

Optimizing humanization strategies involves several considerations:

  • CDR grafting approaches: Carefully transferring the CDRs from the original murine antibody to a human framework while preserving critical framework residues that support CDR conformation

  • Framework selection: Choosing human frameworks with high sequence similarity to the murine framework to minimize structural disruptions

  • Variant generation: Creating multiple variants with different framework modifications (as demonstrated with the five anti-CD11d variants created from the murine 217L clone)

  • Functional screening: Comprehensive screening of humanized variants for:

    • Target binding (flow cytometry, BLI)

    • Neutralization/functional activity (in vitro assays)

    • Lack of off-target binding

    • Absence of unwanted signaling induction

    • Stability and developability properties

  • In vivo validation: Confirming that humanized antibodies retain the therapeutic function of the parental antibody in relevant animal models

What techniques can assess whether antibody binding induces undesired signaling in target cells?

Assessing whether antibody binding induces undesired signaling is critical for therapeutic applications. Researchers can employ several techniques:

  • Phosphorylation studies: Western blotting for phosphorylated signaling proteins (e.g., ERK, AKT, NF-κB) following antibody treatment of target cells

  • Calcium flux assays: Monitoring intracellular calcium levels as an indicator of cellular activation

  • Cytokine secretion: Measuring pro-inflammatory cytokine release following antibody binding

  • Gene expression analysis: RNA-seq or qPCR to detect transcriptional changes associated with cellular activation

  • Functional assays: Assessing changes in cellular behaviors such as migration, adhesion, or respiratory burst

For example, studies with the humanized anti-CD11d-2 clone demonstrated that it binds CD11d/CD18 without inducing inflammatory cell signaling, making it potentially valuable for therapeutic applications where neutralization without activation is desired .

How do CD11d antibodies compare with other therapeutic approaches for inflammatory conditions?

CD11d-targeting antibodies offer several advantages compared to other anti-inflammatory approaches:

  • Specificity: They target a specific integrin primarily expressed on leukocyte subsets involved in inflammation, potentially reducing off-target effects compared to broad-spectrum immunosuppressants

  • Mechanism: By inhibiting leukocyte extravasation into inflamed tissues, these antibodies address a root cause of tissue damage rather than just suppressing symptoms

  • Timing flexibility: Evidence suggests efficacy when administered both prophylactically and therapeutically, allowing flexibility in treatment timing depending on the clinical scenario

  • Compatibility: Potential for combination with other therapeutic agents targeting different mechanisms, as demonstrated by the concept of combining antibodies with non-overlapping epitopes to increase efficacy and decrease escape mutant probability

Compared to small molecule approaches, antibodies typically offer higher specificity and longer half-lives, though they face challenges related to tissue penetration and administration routes .

What are the key considerations for translating CD11d antibody research from animal models to clinical applications?

Translating CD11d antibody therapies to clinical applications requires addressing several critical factors:

  • Species cross-reactivity: Ensuring the humanized antibody maintains binding to human CD11d while demonstrating efficacy in animal models (which may have different CD11d structures)

  • Dosing regimens: Determining optimal dosing based on animal studies while accounting for differences in antibody half-life and target expression between species

  • Safety assessment: Evaluating potential immunogenicity, off-target effects, and the impact of CD11d blockade on normal immune function

  • Indication selection: Identifying the most appropriate inflammatory conditions for initial clinical testing based on preclinical efficacy and understanding of disease pathophysiology

  • Biomarker development: Establishing reliable biomarkers to monitor target engagement and therapeutic response in clinical settings

  • Manufacturing considerations: Ensuring consistent production of antibodies with appropriate post-translational modifications and stability profiles

The successful development of humanized anti-CD11d antibodies represents a significant step toward clinical translation, as these antibodies more closely resemble potential therapeutic agents than their murine counterparts .

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.