CRR6 Antibody

Shipped with Ice Packs
In Stock

Description

Molecular Targeting and Mechanism

CCR6 antibodies bind specifically to extracellular domains of the CCR6 receptor, primarily interfering with its interaction with chemokine ligand CCL20 . Key functional impacts include:

  • Immune cell migration blockade: Inhibits chemotaxis of CCR6-expressing cells (Th17, Treg, B cells) to inflammatory sites

  • Signaling modulation: Demonstrates biased antagonism by selectively blocking β-arrestin recruitment over calcium mobilization

  • Cytokine regulation: Reduces IL-17A production in Th17 cells while preserving regulatory T cell function

Key Research Findings

Recent studies highlight both therapeutic potential and technical challenges:

Table 1: Experimental Antibody Performance

Antibody CloneTarget DomainIC₅₀ (β-arrestin)Migration InhibitionIL-17A ReductionSource
1C6N-terminal10.23 nM60-75%>50%
EPR22259Extracellular loopN/ANot testedNot reported
R6H1Conformational epitopeN/A45-60%Not tested

Notable findings:

  • 1C6 antibody shows dual functionality in blocking cell migration (EC₅₀ = 8.7 nM) and suppressing IL-17A expression at mRNA level

  • EPR22259 demonstrates high specificity in immunohistochemistry (IHC) with <5% cross-reactivity across human tissue microarrays

  • R6H1 enables flow cytometric detection of CCR6 on 15-20% of human PBMCs, correlating with disease activity in psoriasis models

Therapeutic Development Challenges

Current barriers to clinical translation include:

  1. Epitope accessibility: Only 23% of CCR6 surface residues are solvent-accessible, limiting antibody binding sites

  2. Functional selectivity: Most antibodies show signaling bias (e.g., 1C6 affects β-arrestin but not G-protein pathways)

  3. Manufacturing complexity: Requires mammalian expression systems for proper glycosylation (∼4.2 kDa glycan mass)

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Components: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 Weeks (Made-to-Order)
Synonyms
CRR6 antibody; At2g47910 antibody; Protein CHLORORESPIRATORY REDUCTION 6 antibody; chloroplastic antibody
Target Names
CRR6
Uniprot No.

Target Background

Function
This antibody targets a protein essential for both the formation and activity of the chloroplast NAD(P)H dehydrogenase (NDH) complex, a key component of the photosynthetic electron transport chain. Its function may involve the assembly or stabilization of the NDH complex. Specifically, it is required for the accumulation of NDH subcomplex A, a core structural element of the NDH complex. The protein may also participate in post-translational modifications during the biogenesis of subcomplex A.
Gene References Into Functions
  • Studies have demonstrated impaired accumulation of NDH subcomplex A in crr6 and crr7-1 mutants. PMID: 20444231
Database Links

KEGG: ath:AT2G47910

STRING: 3702.AT2G47910.1

UniGene: At.20688

Subcellular Location
Plastid, chloroplast stroma.

Q&A

What is CCR6 and what biological role does it play?

CCR6 is a G protein-coupled receptor (GPCR) involved in a wide range of biological processes. When CCR6 binds to its sole ligand CCL20, it activates a signaling network implicated in immune homeostasis and activation . CCR6 is significantly expressed on several immune cell types, including B lymphocytes, effector and memory T cells, regulatory T cells, and immature dendritic cells . The CCR6/CCL20 axis plays a fundamental role in immune regulation and the recruitment of pro-inflammatory cells to local tissues. This mechanism is particularly important for Th17 cells, which express CCR6 and produce inflammatory cytokines including IL-17, IL-21, and IL-22 .

Why is CCR6 considered a therapeutic target?

CCR6 is considered a valuable therapeutic target because of its involvement in the pathogenesis of multiple diseases. The CCR6/CCL20 pathway is implicated in cancer, psoriasis, multiple sclerosis, HIV infection, and rheumatoid arthritis . Notably, Th17 cells expressing CCR6 release inflammatory cytokines that propagate inflammatory immune responses, making CCR6 antagonists potential treatments for inflammatory diseases like psoriasis or rheumatoid arthritis . Currently, there are no approved drugs targeting CCR6, and development of small molecules against this receptor has proven challenging due to screening difficulties, highlighting the importance of antibody-based approaches .

How does CCR6 contribute to inflammatory disease mechanisms?

CCR6 contributes to inflammatory disease mechanisms primarily through its role in immune cell trafficking and activation. The CCL20/CCR6 axis is crucial for recruiting pro-inflammatory cells, particularly Th17 cells, to sites of inflammation . These Th17 cells produce inflammatory cytokines such as IL-17, which propagate and amplify inflammatory responses. In inflammatory conditions like rheumatoid arthritis and psoriasis, this pathway becomes dysregulated, leading to excessive inflammation and tissue damage. By mediating the migration of these pro-inflammatory cells to local tissues, CCR6 serves as a critical link in the inflammatory cascade, making it a promising target for therapeutic intervention .

What strategies are used to develop monoclonal antibodies against CCR6?

Developing monoclonal antibodies against CCR6 presents unique challenges, as GPCRs are often expressed at low levels and are unstable when purified. One successful approach involves whole cell immunization with cells overexpressing the target receptor. For example, researchers have successfully immunized Balb/c mice with human CCR6 (hCCR6) overexpressing cells to generate monoclonal antibodies . Another approach uses KLH-conjugated peptides from specific regions of CCR6, such as the N-terminal peptide, for immunization . These strategies overcome the difficulties of generating suitable antigens for GPCRs, which require homogenous, stable formats that contain relevant post-translational modifications .

How are anti-CCR6 antibodies validated for research applications?

Validation of anti-CCR6 antibodies involves multiple complementary techniques. Initially, antibodies are tested by flow cytometry for specific binding to CCR6-expressing cells. Further validation includes ELISA assays to determine binding characteristics and epitope mapping . Functional validation is critical and typically includes:

  • β-arrestin recruitment assays to assess the antibody's ability to block CCR6 signaling

  • Calcium mobilization assays to evaluate effects on calcium signaling

  • Chemotaxis assays to determine if the antibody reduces migration of CCR6-expressing cells toward CCL20

  • Expression analysis of downstream inflammatory mediators (e.g., IL-17A in Th17 cells)

Enhanced validation methods may also include immunocytochemistry-immunofluorescence (ICC-IF) and immunohistochemistry (IHC) to confirm tissue specificity and subcellular localization .

What techniques are used to identify the binding epitope of anti-CCR6 antibodies?

Epitope identification for anti-CCR6 antibodies typically employs complementary approaches for thorough characterization. The primary techniques include:

  • Enzyme-linked immunosorbent assay (ELISA) using alanine-substituted peptides: Researchers synthesize point-mutated peptides where each amino acid in the suspected binding region is individually replaced with alanine. Loss of antibody binding to specific mutants identifies critical amino acids within the epitope .

  • Surface plasmon resonance (SPR) analysis: This technique measures the binding kinetics between antibodies and peptides, including association rate (ka), dissociation rate (kd), and dissociation constant (KD). When binding to certain mutants cannot be determined, those amino acids are identified as critical components of the epitope .

For instance, the binding epitope of the anti-mouse CCR6 antibody C6Mab-13 was determined to include amino acids Gly9 and Asp11, with particular importance of Asp11 as demonstrated through both ELISA and SPR methods .

What are the key parameters to measure when characterizing anti-CCR6 antibody binding properties?

When characterizing anti-CCR6 antibody binding properties, several key parameters should be measured:

For example, SPR analysis of the anti-mouse CCR6 antibody C6Mab-13 binding to wild-type peptide showed ka = 6.84 × 10³/ms, kd = 3.77 × 10⁻³/s, and KD = 5.52 × 10⁻⁷ M, with binding completely abolished when Gly9 or Asp11 were mutated to alanine .

How can anti-CCR6 antibodies be used to study inflammatory signaling pathways?

Anti-CCR6 antibodies can be powerful tools for investigating inflammatory signaling pathways through multiple experimental approaches:

  • Signaling pathway dissection: Antagonistic antibodies can be used to block specific aspects of CCR6 signaling. For example, some antibodies selectively inhibit β-arrestin recruitment without affecting calcium mobilization, enabling researchers to study biased signaling mechanisms .

  • Cell migration studies: Chemotaxis assays using anti-CCR6 antibodies can reveal the role of CCR6 in directing immune cell trafficking to inflammatory sites. This helps establish how the CCR6-CCL20 axis contributes to disease pathogenesis .

  • Cytokine expression analysis: Treatment of Th17 cells with anti-CCR6 antibodies followed by RT-qPCR analysis can demonstrate effects on inflammatory cytokine production, particularly IL-17A. This connects CCR6 signaling to downstream inflammatory mediators .

  • In vivo models: Anti-CCR6 antibodies can be administered in animal models of inflammatory diseases to evaluate their potential as therapeutic agents and simultaneously elucidate the role of CCR6 in disease progression .

These applications collectively provide insights into how CCR6 contributes to inflammatory processes and may reveal new therapeutic opportunities.

What experimental controls should be included when using anti-CCR6 antibodies in flow cytometry?

When using anti-CCR6 antibodies in flow cytometry, several critical controls should be included to ensure reliable and interpretable results:

  • Isotype control: Include an antibody of the same isotype (e.g., rat IgG₁ for C6Mab-13) but with irrelevant specificity to detect non-specific binding and establish appropriate gating strategies .

  • Negative cell population: Include cells known not to express CCR6 to establish background staining levels.

  • Positive cell population: Use cells with confirmed CCR6 expression (e.g., Th17 cells or transfected cell lines overexpressing CCR6) as positive controls .

  • Blocking control: Pre-incubate some samples with unlabeled anti-CCR6 antibody or CCR6 peptide to demonstrate specificity through competitive inhibition.

  • Secondary antibody-only control: When using indirect staining methods, include samples with secondary antibody only to assess background from the detection system.

  • FMO (Fluorescence Minus One) controls: Particularly important in multicolor panels to establish proper compensation and gating strategies.

These controls help distinguish true CCR6 expression from technical artifacts and non-specific binding, especially important when studying primary cells with variable receptor expression levels.

How do different anti-CCR6 antibodies affect receptor signaling pathways?

Anti-CCR6 antibodies can differentially affect receptor signaling pathways, demonstrating biased antagonism that targets specific downstream pathways while sparing others. For example, monoclonal antibody 1C6 was found to block β-arrestin recruitment with an IC₅₀ of 10.23 nM but did not inhibit calcium mobilization . This selective inhibition pattern reveals the complexity of GPCR signaling and offers opportunities to target specific disease-relevant pathways.

The differential effects of antibodies on signaling pathways can be evaluated through multiple assays:

  • β-arrestin recruitment assays using GPCR-β-galactosidase fusion proteins to measure recruitment following CCL20 stimulation with and without antibody treatment .

  • Calcium mobilization assays to assess effects on G protein-dependent signaling using fluorescent calcium indicators or enzymatic readouts .

  • Chemotaxis assays to evaluate functional consequences on cell migration toward CCL20 .

Understanding these differential effects is important for both basic research into GPCR signaling mechanisms and therapeutic antibody development, as it allows for targeting specific disease-relevant pathways while potentially minimizing unwanted effects.

What techniques are most effective for assessing anti-CCR6 antibody effects on immune cell migration?

Several complementary techniques effectively assess anti-CCR6 antibody effects on immune cell migration:

  • Transwell chemotaxis assays: Cells expressing CCR6 are placed in the upper chamber of a Transwell system with CCL20 in the lower chamber. Anti-CCR6 antibodies are added to test their ability to inhibit migration. This quantitative method provides direct measurement of migration inhibition and allows for dose-response analyses .

  • Real-time cell migration tracking: Live-cell imaging systems track individual cell movements in response to chemokine gradients with and without antibody treatment, providing detailed information about migration velocity, directionality, and persistence.

  • 3D matrix migration assays: Embedding cells in collagen or Matrigel creates a more physiologically relevant environment for studying migration through tissues, better mimicking in vivo conditions.

  • In vivo migration assays: Fluorescently labeled cells treated with anti-CCR6 antibodies can be transferred into animals to track their migration to CCL20-expressing tissues, providing the most physiologically relevant assessment.

The choice of technique depends on the specific research question, with transwell assays offering simplicity and quantitative readouts, while more complex systems provide greater physiological relevance.

How can anti-CCR6 antibodies be used to distinguish different T cell subsets in heterogeneous samples?

Anti-CCR6 antibodies can be valuable tools for distinguishing T cell subsets in heterogeneous samples through multi-parameter flow cytometry and other techniques. CCR6 serves as a particularly useful marker because of its differential expression across T cell subpopulations, especially for identifying Th17 cells . To effectively distinguish T cell subsets:

  • Combine anti-CCR6 antibodies with antibodies against other lineage-defining markers: For Th17 cells, include markers for IL-17, RORγt, and CD4; for Tregs, include FoxP3 and CD25; and for other helper T cells, include markers like CXCR3 (Th1) and CCR4 (Th2).

  • Use intracellular cytokine staining: After appropriate stimulation, combine CCR6 surface staining with intracellular staining for cytokines like IL-17A to confirm functional Th17 identity .

  • Implement advanced analysis techniques: Utilize high-dimensional analysis methods like t-SNE or UMAP to identify distinct populations in complex datasets from techniques such as mass cytometry or spectral flow cytometry.

  • Validate with functional assays: Confirm the identity of CCR6+ populations through functional assays such as cytokine production profiles or migration responses to CCL20 .

This approach allows researchers to accurately identify and isolate specific T cell subsets from heterogeneous samples for downstream analyses, which is particularly valuable for studying inflammatory and autoimmune conditions.

What are the challenges in developing therapeutic antibodies targeting the CCR6/CCL20 axis?

Developing therapeutic antibodies targeting the CCR6/CCL20 axis faces several significant challenges:

  • GPCR-specific challenges: GPCRs like CCR6 are difficult targets because they are often expressed at low levels, have complex three-dimensional structures with multiple transmembrane domains, and are unstable when purified. These characteristics complicate the generation of suitable antigens for antibody development .

  • Epitope accessibility: The extracellular domains of CCR6 that are accessible to antibodies are relatively small, limiting potential binding sites. The N-terminal domain and extracellular loops represent the main targetable regions .

  • Functional antagonism: Developing antibodies that not only bind to CCR6 but also functionally block its interaction with CCL20 or inhibit downstream signaling requires extensive screening and characterization .

  • Signaling complexity: CCR6 activates multiple signaling pathways, and therapeutic antibodies may affect these pathways differently (biased antagonism). Understanding which pathways are most relevant for specific diseases is challenging .

  • Species differences: Significant differences between human and mouse CCR6 can complicate the translation of findings from animal models to human applications, requiring species-specific antibodies for preclinical development .

  • Tissue penetration: As large molecules, antibodies may have limited access to certain tissues, potentially restricting their efficacy in some disease contexts.

Addressing these challenges requires innovative approaches in antibody engineering, screening methods, and translational models to develop effective therapeutic candidates.

How can epitope mapping data guide the development of more effective anti-CCR6 antibodies?

Epitope mapping data can significantly guide the development of more effective anti-CCR6 antibodies through several strategic approaches:

  • Structure-function correlation: Identifying critical binding residues, such as Gly9 and Asp11 in mouse CCR6, provides insights into functional domains of the receptor . Antibodies targeting epitopes near the CCL20 binding site may more effectively block ligand interaction, while those targeting other regions might preferentially affect receptor conformation or dimerization.

  • Rational antibody engineering: With precise epitope knowledge, researchers can modify antibodies to enhance their binding properties or functional effects. This may include affinity maturation focused on interactions with key residues or engineering bispecific antibodies that simultaneously target multiple critical epitopes.

  • Species cross-reactivity prediction: Comparing epitope sequences between species helps predict whether antibodies will cross-react. For instance, if the epitope identified in mouse CCR6 is conserved in human CCR6, the antibody may have translational potential .

  • Mechanism of action insights: The binding epitope often correlates with the antibody's functional effects. For example, antibodies binding to the N-terminal domain may have different signaling effects than those binding to extracellular loops, potentially explaining biased antagonism phenomena .

  • Immunogenicity assessment: Epitope mapping helps identify antibodies targeting highly conserved regions, which may be less immunogenic and therefore more suitable for therapeutic development.

By systematically applying epitope mapping data, researchers can design next-generation anti-CCR6 antibodies with improved specificity, potency, and therapeutic efficacy for treating inflammatory and autoimmune diseases.

What considerations are important when interpreting conflicting data from different anti-CCR6 antibody clones?

When interpreting conflicting data from different anti-CCR6 antibody clones, researchers should consider several important factors:

When faced with conflicting data, researchers should systematically evaluate these factors and consider conducting side-by-side comparisons of different antibody clones using standardized protocols and multiple complementary assays.

How might anti-CCR6 antibodies be utilized in immunotherapy approaches?

Anti-CCR6 antibodies hold significant potential for various immunotherapy approaches, particularly for conditions where the CCR6/CCL20 axis drives pathogenesis:

  • Inflammatory disease treatment: Anti-CCR6 antibodies could selectively inhibit the migration of pro-inflammatory Th17 cells to affected tissues in conditions like psoriasis and rheumatoid arthritis, reducing local inflammation without broad immunosuppression .

  • Combination immunotherapy: Anti-CCR6 antibodies might be combined with existing immunotherapies to enhance efficacy. For example, in autoimmune conditions, combining CCR6 blockade with cytokine-targeting biologics could simultaneously inhibit multiple inflammatory pathways.

  • Cancer immunotherapy: Since CCR6 is implicated in certain cancers, anti-CCR6 antibodies could potentially modulate the tumor microenvironment by altering immune cell recruitment or directly affecting CCR6-expressing tumor cells .

  • Targeted immune cell depletion: Antibody-drug conjugates targeting CCR6 could selectively eliminate pathogenic Th17 cells in autoimmune diseases while sparing other T cell populations necessary for normal immune function.

  • Modulation of antibody-dependent cellular cytotoxicity (ADCC): Engineering anti-CCR6 antibodies with enhanced Fc effector functions could promote selective depletion of CCR6-expressing cells through ADCC mechanisms.

These approaches leverage the specificity of anti-CCR6 antibodies to modulate immune responses in a targeted manner, potentially offering improved therapeutic options with fewer side effects compared to broad immunosuppression.

What emerging technologies are improving anti-CCR6 antibody development and characterization?

Several emerging technologies are significantly enhancing anti-CCR6 antibody development and characterization:

  • Single B cell antibody discovery: This technology isolates individual B cells from immunized animals, sequences their antibody genes, and recombinantly expresses the antibodies. This approach captures a broader antibody repertoire than traditional hybridoma methods, potentially yielding more diverse anti-CCR6 antibodies with unique properties .

  • Phage display with synthetic libraries: Advanced phage display libraries allow screening against specific CCR6 epitopes or conformations, enabling the identification of antibodies with tailored binding properties without animal immunization.

  • Cryo-electron microscopy (cryo-EM): This technique can provide high-resolution structural information about antibody-CCR6 complexes, offering unprecedented insights into binding mechanisms and conformational changes upon antibody binding.

  • Surface plasmon resonance (SPR) combined with hydrogen-deuterium exchange mass spectrometry (HDX-MS): This combination provides detailed information about both binding kinetics and conformational changes induced by antibody binding, helping elucidate mechanisms of action .

  • CRISPR/Cas9 genome editing: Creating precisely engineered cell lines with modified CCR6 variants helps validate antibody specificity and epitope recognition, supporting more rigorous characterization.

  • Artificial intelligence for antibody design: Machine learning approaches can predict optimal antibody sequences based on epitope information, potentially accelerating the development of high-affinity, functionally active anti-CCR6 antibodies.

These technologies collectively advance our ability to develop well-characterized, functionally optimized anti-CCR6 antibodies for both research and therapeutic applications.

How can long-term stability and functionality of anti-CCR6 antibodies be optimized for research applications?

Optimizing the long-term stability and functionality of anti-CCR6 antibodies for research applications requires attention to several key factors:

  • Buffer optimization: Developing ideal storage buffers that maintain antibody stability is critical. Typically, these include:

    • Appropriate pH (usually 7.2-7.4)

    • Protein stabilizers (e.g., BSA or gelatin at 0.1-1%)

    • Mild preservatives (e.g., 0.02% sodium azide)

    • Cryoprotectants for freeze-thaw stability (e.g., glycerol at 10-50%)

  • Storage conditions: Store antibodies at recommended temperatures, typically -20°C or -80°C for long-term storage, with working aliquots at 4°C to minimize freeze-thaw cycles that can lead to aggregation and loss of activity.

  • Formulation strategies: Consider lyophilization for extended shelf-life, particularly for distributing antibodies to multiple research labs. Lyophilized antibodies are more stable at ambient temperatures and during shipping.

  • Quality control protocols: Implement regular testing of antibody functionality using:

    • Binding assays (ELISA, flow cytometry) to confirm target recognition is maintained

    • Functional assays (e.g., β-arrestin recruitment inhibition or chemotaxis assays) to verify biological activity

    • Physicochemical assessments (size exclusion chromatography, dynamic light scattering) to detect aggregation

  • Antibody engineering: For particularly valuable research antibodies, consider stability-enhancing modifications such as:

    • Framework mutations that increase thermostability

    • Removal of unpaired cysteines to prevent disulfide scrambling

    • Glycoengineering to optimize glycosylation profiles

By systematically addressing these aspects, researchers can ensure that anti-CCR6 antibodies maintain their specific binding properties and functional characteristics throughout long-term research applications, enhancing experimental reproducibility and reliability.

How do anti-CCR6 antibody studies in animal models translate to human inflammatory conditions?

Translating anti-CCR6 antibody studies from animal models to human inflammatory conditions requires careful consideration of several factors:

Successful translation typically involves iterative refinement, starting with mechanistic studies in mouse models, progressing to ex vivo human cell systems, and finally advancing to clinical trials with humanized or fully human antibodies that target epitopes conserved between species.

What biomarkers can be used to monitor anti-CCR6 antibody efficacy in experimental systems?

Several biomarkers can effectively monitor anti-CCR6 antibody efficacy in experimental systems:

  • Direct target engagement markers:

    • CCR6 receptor occupancy by flow cytometry, measuring the binding of fluorescently labeled CCL20 or a non-competing anti-CCR6 antibody before and after treatment

    • Reduction in CCL20-induced calcium flux in target cells

    • Inhibition of β-arrestin recruitment following CCL20 stimulation

  • Functional response markers:

    • Decreased chemotaxis of CCR6+ cells toward CCL20 in migration assays

    • Reduced infiltration of CCR6+ cells in tissue samples from animal models

    • Changes in the distribution of CCR6+ cell populations in blood vs. tissues

  • Downstream signaling and inflammation markers:

    • Reduced expression of IL-17A and other inflammatory cytokines in Th17 cells, measured by RT-qPCR or protein assays

    • Decreased phosphorylation of signaling proteins downstream of CCR6

    • Changes in inflammatory gene expression profiles in affected tissues

  • Disease-specific markers:

    • For psoriasis models: reduced epidermal thickness, keratinocyte proliferation, and skin inflammation scores

    • For arthritis models: decreased joint swelling, cartilage destruction, and bone erosion

    • For inflammatory bowel disease models: improved histological scores and reduced intestinal permeability

These biomarkers should be selected based on the specific research question and experimental system, ideally combining direct target engagement measures with functional outcomes and disease-relevant endpoints to comprehensively assess antibody efficacy.

How can anti-CCR6 antibodies be used to identify novel therapeutic targets in the CCR6/CCL20 signaling pathway?

Anti-CCR6 antibodies can serve as valuable tools for identifying novel therapeutic targets in the CCR6/CCL20 signaling pathway through several research strategies:

  • Signaling pathway dissection: By selectively blocking CCR6 with antibodies that demonstrate biased antagonism (e.g., inhibiting β-arrestin recruitment without affecting calcium mobilization) , researchers can identify which downstream pathways are critical for specific disease manifestations. This approach can reveal new druggable nodes in the signaling network beyond CCR6 itself.

  • Protein-protein interaction studies: Using anti-CCR6 antibodies for immunoprecipitation followed by mass spectrometry can identify novel interaction partners of CCR6. These interactors might represent previously unrecognized components of the signaling pathway that could serve as alternative therapeutic targets.

  • Transcriptomic and proteomic profiling: Comparing gene and protein expression profiles in CCR6+ cells before and after antibody treatment can reveal:

    • Secondary mediators induced by CCR6 signaling

    • Feedback mechanisms that regulate receptor function

    • Cell type-specific responses to CCR6 blockade

  • Combination therapy studies: Using anti-CCR6 antibodies in combination with inhibitors of other inflammatory pathways can identify synergistic interactions, suggesting rational combination therapies for enhanced efficacy.

  • In vivo imaging: Labeled anti-CCR6 antibodies can be used to track CCR6+ cells in vivo, revealing previously unknown migration patterns or tissue niches that might be targeted therapeutically.

  • Resistance mechanisms: Studying cases where anti-CCR6 antibody treatment is ineffective can uncover compensatory pathways that become activated, identifying additional targets for combination approaches.

By applying these approaches systematically, researchers can move beyond targeting CCR6 directly to develop more refined therapeutic strategies addressing specific aspects of the CCR6/CCL20 signaling network.

What controls and validation steps are essential when developing a new anti-CCR6 antibody?

Developing a new anti-CCR6 antibody requires rigorous controls and validation steps to ensure specificity, functionality, and reliability:

  • Initial screening and selection:

    • Screen antibodies against both CCR6-positive and CCR6-negative cell lines to confirm specificity

    • Test binding to CCR6 from relevant species (human, mouse) depending on intended applications

    • Use competitive binding assays with known ligands (CCL20) to identify antibodies targeting functional epitopes

  • Biochemical characterization:

    • Determine binding affinity, association and dissociation rates using surface plasmon resonance

    • Map the epitope using alanine-scanning mutagenesis or hydrogen-deuterium exchange mass spectrometry

    • Confirm specificity against related chemokine receptors to rule out cross-reactivity

  • Functional validation:

    • Assess effects on receptor signaling through:

      • β-arrestin recruitment assays

      • Calcium mobilization assays

      • Chemotaxis assays

    • Confirm biological activity in relevant primary cells (e.g., Th17 cells) by measuring IL-17 expression

  • Application-specific validation:

    • For flow cytometry: Verify performance across different fixation methods and cell types

    • For immunohistochemistry/immunofluorescence: Validate on both frozen and fixed tissues with appropriate controls

    • For neutralization studies: Establish dose-response relationships for inhibitory effects

  • Reproducibility testing:

    • Test multiple antibody batches to ensure consistent performance

    • Validate using multiple detection methods and experimental systems

    • Have independent laboratories confirm key findings when possible

These comprehensive validation steps ensure that new anti-CCR6 antibodies will provide reliable and interpretable results across different research applications.

How should researchers optimize immunostaining protocols for detecting CCR6 in tissue samples?

Optimizing immunostaining protocols for detecting CCR6 in tissue samples requires attention to several critical factors:

  • Sample preparation and fixation:

    • Test multiple fixation methods: CCR6 is a GPCR with complex structure, and excessive fixation can mask epitopes. Compare paraformaldehyde (2-4%), methanol, and acetone fixation to determine optimal epitope preservation.

    • Consider antigen retrieval methods: For formalin-fixed paraffin-embedded (FFPE) tissues, test heat-induced epitope retrieval methods using citrate buffer (pH 6.0) or Tris-EDTA buffer (pH 9.0) .

    • Minimize autofluorescence: For fluorescent detection, use Sudan Black B or commercial autofluorescence quenchers to reduce background in tissues with high autofluorescence like liver or brain.

  • Blocking and antibody incubation:

    • Implement thorough blocking: Use species-appropriate serum (5-10%) with added detergent (0.1-0.3% Triton X-100) to reduce non-specific binding.

    • Optimize antibody concentration: Perform titration experiments (typically 0.1-10 μg/ml) to determine the optimal concentration that maximizes specific signal while minimizing background .

    • Extend incubation times: Consider overnight incubation at 4°C for primary antibodies to improve signal-to-noise ratio, particularly for low-abundance receptors like CCR6.

  • Detection systems:

    • Compare direct vs. indirect detection: While direct conjugates offer simplicity, indirect methods (secondary antibodies) often provide signal amplification beneficial for CCR6 detection.

    • Consider signal amplification: For tissues with low CCR6 expression, tyramide signal amplification or polymer-based detection systems can enhance sensitivity.

    • Test counterstains: Select nuclear counterstains (DAPI, Hoechst) and additional markers that complement CCR6 detection without spectrum overlap.

  • Essential controls:

    • Include negative controls: Isotype controls at the same concentration as the primary antibody are essential .

    • Use positive controls: Include known CCR6-positive tissues (e.g., lymphoid tissues) or CCR6-transfected cell pellets embedded alongside test samples.

    • Perform peptide competition: Pre-incubation of the antibody with the immunizing peptide should abolish specific staining .

    • Include knockout/knockdown validation when available: Tissue from CCR6-knockout animals provides the gold standard negative control.

By systematically optimizing these parameters, researchers can develop robust immunostaining protocols for reliable CCR6 detection in tissue samples.

What factors influence the selection of anti-CCR6 antibodies for specific research applications?

Selecting the appropriate anti-CCR6 antibody for specific research applications requires careful consideration of multiple factors:

  • Application-specific requirements:

    • Flow cytometry: Prefer antibodies validated specifically for flow cytometry, with bright fluorophores appropriate for the target cell population's expression level .

    • Immunohistochemistry: Select antibodies validated with enhanced validation for IHC, considering whether they work on frozen sections, FFPE tissues, or both .

    • Functional studies: Choose antibodies characterized for neutralizing activity with established IC₅₀ values in relevant functional assays .

  • Epitope considerations:

    • Epitope location: Antibodies targeting different domains (N-terminus, extracellular loops) may perform differently across applications .

    • Conformational sensitivity: Some antibodies recognize only native protein conformations, limiting their use in applications involving denaturation.

    • Species cross-reactivity: If working across species, confirm whether the epitope is conserved and whether the antibody has been validated in all relevant species .

  • Technical characteristics:

    • Clonality: Monoclonal antibodies offer consistency and specificity for defined epitopes, while polyclonal antibodies may provide enhanced signal through multiple epitope recognition .

    • Isotype: Consider the isotype (IgG₁, IgG₂, etc.) based on secondary detection systems and potential for unwanted Fc receptor interactions.

    • Affinity and specificity: Higher affinity antibodies generally perform better in applications detecting low-abundance targets, but extremely high affinity may increase background in some contexts.

  • Validation depth:

    • Enhanced validation: Prefer antibodies validated through multiple orthogonal methods appropriate for your application .

    • Independent validation: Consider whether the antibody has been validated by multiple laboratories or only by the manufacturer.

    • Publication record: Antibodies with successful use in peer-reviewed publications for your specific application offer greater confidence.

  • Experimental goals:

    • Descriptive studies: Antibodies with high sensitivity may be prioritized for detection of all CCR6+ cells.

    • Functional studies: Antibodies with well-characterized effects on receptor signaling are essential .

    • Quantitative analysis: Antibodies with linear signal-to-expression relationships are preferred for quantitative applications.

By systematically evaluating these factors, researchers can select anti-CCR6 antibodies optimally suited to their specific experimental requirements, enhancing the reliability and interpretability of their results.

How might single-cell analysis techniques enhance our understanding of CCR6 biology using anti-CCR6 antibodies?

Single-cell analysis techniques combined with anti-CCR6 antibodies offer powerful approaches to uncover new insights into CCR6 biology:

  • Single-cell RNA sequencing (scRNA-seq) with antibody-based cell sorting:

    • Anti-CCR6 antibodies can isolate pure populations of CCR6+ cells for scRNA-seq analysis, revealing heterogeneity within seemingly uniform CCR6+ populations like Th17 cells .

    • This approach can identify previously unrecognized CCR6+ cell subsets with distinct transcriptional programs and potentially different roles in disease processes.

    • Comparing CCR6+ cells from healthy and diseased tissues can uncover disease-specific alterations in gene expression patterns.

  • Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq):

    • This technique simultaneously quantifies surface protein expression (including CCR6) and the transcriptome at single-cell resolution.

    • CITE-seq can reveal relationships between CCR6 protein levels and transcriptional states, potentially identifying cells transitioning between phenotypes during immune responses.

    • By incorporating multiple antibodies, researchers can map CCR6 co-expression patterns with other chemokine receptors and immune markers at unprecedented resolution.

  • Single-cell proteomics and phospho-flow cytometry:

    • These approaches can track CCR6-mediated signaling events at the single-cell level, revealing how individual cells respond to CCL20 stimulation.

    • Anti-CCR6 antibodies combined with phospho-specific antibodies can identify which signaling pathways are activated in specific cell subsets.

    • This can uncover signaling heterogeneity that may explain differential responses to therapeutic CCR6 blockade.

  • Spatial transcriptomics and multiplexed imaging:

    • Combining anti-CCR6 antibodies with spatial transcriptomics or multiplexed imaging techniques (e.g., Imaging Mass Cytometry, CODEX) can map the location of CCR6+ cells within tissues in relation to other cell types and anatomical structures .

    • This reveals spatial relationships that may be crucial for understanding CCR6 function in complex tissue environments during inflammation.

These single-cell approaches can transform our understanding of CCR6 biology by uncovering functional heterogeneity, revealing rare cell populations, and mapping complex cellular interactions in inflammatory and autoimmune diseases.

What is the potential for developing bispecific antibodies targeting CCR6 and other inflammatory markers?

Bispecific antibodies targeting CCR6 and other inflammatory markers hold substantial potential for enhancing therapeutic efficacy through several mechanisms:

  • Dual targeting strategies:

    • CCR6/IL-17 bispecifics: By simultaneously targeting CCR6 on Th17 cells and neutralizing their key effector cytokine IL-17, these antibodies could provide synergistic inhibition of inflammatory cascades in conditions like psoriasis .

    • CCR6/CCR4 bispecifics: Since many inflammatory T cells co-express multiple chemokine receptors, dual blockade could more effectively prevent cellular trafficking to inflammatory sites than targeting either receptor alone.

    • CCR6/TNF-α bispecifics: Combining blockade of CCR6-mediated cell recruitment with neutralization of a master inflammatory cytokine could address multiple aspects of inflammatory diseases simultaneously.

  • Enhanced targeting specificity:

    • By requiring engagement of both CCR6 and a second marker, bispecific antibodies can increase cellular specificity, potentially reducing off-target effects compared to conventional CCR6 antibodies.

    • This approach could selectively target pathogenic CCR6+ cells (e.g., those also expressing activation markers) while sparing regulatory CCR6+ populations.

  • Novel functional mechanisms:

    • Selective cell depletion: Bispecifics targeting CCR6 and CD3 could redirect T cell cytotoxicity against CCR6-high pathogenic cells in autoimmune diseases.

    • Immune checkpoint modulation: CCR6/PD-1 or CCR6/CTLA-4 bispecifics could simultaneously block inflammatory cell trafficking while enhancing coinhibitory signals to dampen pathogenic immune responses.

    • Targeted cytokine delivery: CCR6-targeted bispecifics carrying an anti-inflammatory cytokine domain (e.g., IL-10) could deliver immunomodulatory signals specifically to CCR6+ cells.

  • Technical advantages:

    • Simplified treatment regimens compared to combination therapy with two separate antibodies

    • Potential for improved pharmacokinetics and tissue penetration through novel antibody formats

    • Possibility of creating novel functionalities not achievable with monospecific antibodies

The development of such bispecific antibodies faces challenges including optimal format selection, maintaining dual-binding capacity, and complex manufacturing, but represents a promising frontier for next-generation therapeutics targeting CCR6-mediated inflammatory diseases.

How might artificial intelligence and machine learning approaches enhance anti-CCR6 antibody development and research?

Artificial intelligence (AI) and machine learning (ML) approaches offer transformative potential for enhancing anti-CCR6 antibody development and research through several innovative applications:

  • Epitope prediction and antibody design:

    • AI algorithms can analyze CCR6 protein structure to predict optimal epitopes for antibody targeting, particularly focusing on regions critical for CCL20 binding or receptor activation.

    • Machine learning models trained on antibody-antigen interaction data can design optimized antibody sequences with enhanced affinity, specificity, or functional properties for targeting CCR6.

    • These approaches could identify novel epitopes beyond those traditionally targeted, potentially yielding antibodies with unique functional properties .

  • Image analysis and phenotyping:

    • Deep learning algorithms can analyze immunohistochemistry or immunofluorescence images to quantify CCR6 expression patterns in tissues with greater accuracy and reproducibility than manual assessment .

    • AI-powered flow cytometry analysis can identify subtle CCR6+ cell populations in complex samples that might be missed with conventional gating strategies.

    • Machine learning can integrate CCR6 expression data with other markers to develop improved classification systems for immune cell subsets in health and disease.

  • Predictive modeling for therapeutic applications:

    • AI can analyze comprehensive datasets to predict which patients might respond best to CCR6-targeted therapies based on biomarker profiles.

    • Machine learning models can predict potential off-target effects or cross-reactivity of anti-CCR6 antibodies before experimental testing.

    • Computational models can simulate the effects of different antibody binding modes on CCR6 signaling pathways, helping select candidates with optimal functional profiles .

  • Drug discovery acceleration:

    • High-throughput screening data from anti-CCR6 antibody candidates can be analyzed with AI to identify subtle patterns correlating molecular features with functional outcomes.

    • Virtual screening approaches can pre-select promising antibody candidates before experimental testing, reducing development time and costs.

    • Machine learning can optimize antibody manufacturing conditions by predicting how process parameters affect antibody quality and yield.

  • Literature mining and knowledge integration:

    • Natural language processing can extract and synthesize information about CCR6 across thousands of research papers, identifying connections and insights that might be missed by human researchers.

    • AI systems can integrate diverse data types (genomic, proteomic, clinical) to generate new hypotheses about CCR6 biology and potential therapeutic applications.

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.