ACR4 (Arabidopsis CRINKLY 4) is a receptor-like kinase (RLK) critical for regulating cell division, differentiation, and tissue organization in plants, particularly in Arabidopsis thaliana. It is involved in maintaining L1 cell layer integrity, root meristem development, and ovule integument formation . ACR4’s intracellular domain (ICD) undergoes autophosphorylation and interacts with proteins such as PROTEIN PHOSPHATASE 2A-3 (PP2A-3) and transcription factors like WOX5, which regulate formative cell divisions .
While the term “ACR4 antibody” is not explicitly defined in the provided literature, antibodies targeting analogous receptors (e.g., CXCR4 or ACKR4) are well-documented. For example:
Anti-CXCR4 antibodies (e.g., ACR-014-AG) are used to study chemokine receptor CXCR4 in humans, rats, and mice .
Anti-ACKR4 monoclonal antibodies (e.g., A4Mab-1, A4Mab-2) detect mouse atypical chemokine receptor 4 (ACKR4) in flow cytometry and western blotting .
These antibodies are distinct from plant ACR4 but share nomenclature similarities.
Phosphorylation and Binding Partners:
Structural Features:
For mammalian receptors like ACKR4:
Antibody Validation:
Epitope Mapping:
No studies in the provided sources describe antibodies directly targeting plant ACR4. Research on ACR4 has focused on genetic and biochemical interactions rather than immunodetection. Future work could explore:
Development of polyclonal/monoclonal antibodies against ACR4 epitopes.
Applications in plant tissue imaging or protein localization studies.
ACR4 refers to two distinct proteins in scientific literature: Arabidopsis CRINKLY4 in plant biology and Atypical Chemokine Receptor 4 in mammalian systems. In plants, Arabidopsis CRINKLY4 (ACR4) functions as a receptor-like kinase (RLK) involved in global plant development, with particular importance in formative cell division processes . In mammalian systems, Atypical Chemokine Receptor 4 (ACKR4) regulates dendritic cell migration by controlling chemokine ligands, notably binding to CCR7 ligands such as CCL19 and CCL21, and is involved in tumor development in mouse models . Unlike typical chemokine receptors, ACKR4 does not induce classical G protein-coupled receptor signaling but instead facilitates chemokine degradation through β-arrestin-mediated endocytosis and lysosomal processing .
Multiple complementary approaches have proven effective for studying ACR4 protein interactions. For plant ACR4, researchers have successfully employed in silico analysis, tandem affinity purification (TAP), yeast two-hybrid (Y2H) assays, and phage display approaches to define ACR4-interacting proteins . For instance, TAP approaches with Arabidopsis cell suspension cultures expressing tagged ACR4 intracellular kinase domains identified potential interacting proteins including HTPA REDUCTASE 1/DAPB1, HTPA REDUCTASE 2/DAPB2, and PROTEIN PHOSPHATASE 2A-3 (PP2A-3) . In vitro validation of interactions can be performed using gel-filtration analyses and pull-down assays, while in planta confirmation can utilize coimmunoprecipitation in transient expression systems such as Nicotiana benthamiana .
Validating antibody specificity requires a multi-faceted approach. For anti-mouse ACKR4 antibodies, researchers have implemented a systematic validation strategy including:
Initial screening via enzyme-linked immunosorbent assay (ELISA) using the target peptide
Secondary screening by flow cytometry comparing reactivity between receptor-expressing cells (e.g., CHO/mACKR4) and control cells (e.g., CHO-K1)
Specificity confirmation through peptide blocking experiments in western blotting
Quantitative assessment of binding affinity using flow cytometry to determine dissociation constant (KD) values
For example, western blotting validation of anti-mACKR4 antibodies (A4Mab-1 and A4Mab-2) demonstrated specific detection of mACKR4 as a ~50-kDa band in LN229/mACKR4 cell lysates but not in control LN229 cells, with this detection being successfully blocked in the presence of mACKR4 peptide .
Developing antibodies against membrane proteins presents unique challenges due to their complex structure and hydrophobic domains. Based on successful development of anti-mACKR4 antibodies, two primary approaches emerge:
N-terminal peptide immunization strategy: This approach involves:
Selecting immunogenic peptide sequences from the extracellular N-terminal domain
Conjugating the peptide to a carrier protein (e.g., KLH)
Immunizing animals with the peptide-carrier conjugate using an appropriate adjuvant
Screening hybridomas through a two-step process: ELISA against naked peptide followed by flow cytometry to identify clones recognizing the native conformation
Cell-Based Immunization and Screening (CBIS) method: This alternative approach has been successfully employed for developing antibodies against multiple mouse chemokine receptors and involves immunizing with receptor-expressing cells .
For mACKR4, the N-terminal peptide immunization approach successfully yielded three monoclonal antibodies (A4Mab-1, A4Mab-2, and A4Mab-3) with different binding characteristics and applications .
Understanding the binding characteristics of different antibody clones is critical for experimental design. The following table summarizes key properties of three anti-mACKR4 antibody clones:
These differences in binding properties should guide experimental design:
For flow cytometry applications requiring highest sensitivity, A4Mab-3 may be preferred due to its superior affinity
For western blotting applications, A4Mab-2 demonstrates superior reactivity
For multi-method approaches requiring both flow cytometry and western blotting, A4Mab-1 or A4Mab-2 would be more appropriate
Several confounding factors must be considered when investigating ACR4 protein-protein interactions:
Technical limitations with membrane proteins: The transmembrane nature of ACR4 presents challenges for in vitro studies. This has led researchers to focus primarily on intracellular domains for in vitro and in vivo studies .
Limited overlap between detection methods: Different protein interaction detection approaches (e.g., TAP, Y2H, phage display) may yield distinct sets of potential interacting partners with minimal overlap. For example, studies with plant ACR4 found limited to no overlap between interaction partners identified by different methods, suggesting either method-specific detection bias or high false-positive rates .
Expression domain considerations: Potential interacting proteins may show distinct expression patterns that only partially overlap with the ACR4 expression domain, requiring careful consideration of spatiotemporal dynamics .
Post-translational modifications: Phosphorylation can significantly impact protein interactions. For ACR4, the interaction with some partners (like PP2A-3) may be phosphorylation-dependent, requiring careful experimental design to account for the phosphorylation state .
When using ACR4 antibodies for flow cytometry, consider the following protocol optimizations:
Cell preparation:
For adherent cells (e.g., CHO-K1, LN229), harvest cells in exponential growth phase using non-enzymatic cell dissociation solution to preserve surface epitopes
Wash cells twice with 1% bovine serum albumin (BSA) in phosphate-buffered saline (PBS)
Adjust cell concentration to 1 × 10⁶ cells/mL
Antibody incubation:
Incubate 100 μL of cell suspension with optimized antibody concentration (typically 1 μg/mL for A4Mab-1, A4Mab-2, or A4Mab-3)
Maintain incubation for 30 minutes on ice
Wash cells twice with 1% BSA in PBS
Secondary antibody:
For optimal results, always include appropriate isotype controls and positive/negative cell lines to establish gating strategies.
Accurate determination of binding affinities is essential for characterizing antibody performance. For anti-mACKR4 antibodies, researchers employed the following methodology to determine dissociation constant (KD) values:
Sample preparation:
Harvest cells expressing the target receptor (e.g., CHO/mACKR4) in exponential growth phase
Prepare serial dilutions of purified antibody ranging from 100 nM to 0.1 nM
Flow cytometry analysis:
Incubate cells with different antibody concentrations under equilibrium conditions
Detect bound antibody using fluorochrome-conjugated secondary antibody
Measure mean fluorescence intensity (MFI) for each antibody concentration
Data analysis:
Plot MFI versus antibody concentration
Fit data to a one-site binding model using appropriate statistical software
Calculate KD as the antibody concentration yielding half-maximal binding
Using this approach, researchers determined KD values of 6.0 × 10⁻⁹ M, 1.3 × 10⁻⁸ M, and 1.7 × 10⁻⁹ M for A4Mab-1, A4Mab-2, and A4Mab-3, respectively, providing quantitative measures of their binding affinities .
When performing western blotting with ACR4 antibodies, incorporate the following essential controls:
Positive and negative cell lysates:
Loading controls:
Peptide blocking controls:
Molecular weight markers:
Implementation of these controls is exemplified in the development of anti-mACKR4 antibodies, where A4Mab-1 and A4Mab-2 demonstrated specific detection of mACKR4 as a ~50-kDa band, with this detection being blocked in the presence of mACKR4 peptide .
ACR4 antibodies offer significant potential for understanding tumor development mechanisms, particularly through investigation of ACKR4's role in immune cell migration and tumor immunology. ACKR4 regulates dendritic cell migration by controlling chemokine ligands and has been implicated in tumor development in mouse models . Specific anti-mACKR4 antibodies enable:
Identification and characterization of ACKR4-expressing cells in the tumor microenvironment using flow cytometry, potentially revealing novel cellular populations involved in tumor progression or suppression
Quantification of ACKR4 expression levels in different tumor types and stages, possibly identifying correlations with disease progression or treatment response
Investigation of ACKR4's role in chemokine-mediated immune cell trafficking within tumors, potentially revealing mechanisms by which tumors evade immune surveillance
Development of therapeutic strategies targeting ACKR4 to modulate immune responses in cancer, as preliminary findings suggest that targeting ACKR4 might improve immunotherapy efficacy
The availability of well-characterized anti-mACKR4 monoclonal antibodies suitable for flow cytometry and western blotting provides researchers with essential tools to pursue these investigations in preclinical tumor models .
While plant ACR4 (Arabidopsis CRINKLY4) and mammalian ACKR4 (Atypical Chemokine Receptor 4) are distinct proteins with different functions, several methodological parallels in their study may inform cross-disciplinary approaches:
Protein interaction network analysis:
Domain-focused investigations:
Phosphorylation dynamics:
Antibody development strategies:
Despite functional differences, shared methodological challenges and solutions between these research areas could accelerate progress in both fields.
Natural antibodies and laboratory-generated monoclonal antibodies each offer distinct advantages and limitations for research applications:
Natural antibodies (such as those observed in Alzheimer's disease patients against proteins like ankyrin G):
Develop through natural immune responses to endogenous or disease-associated antigens
May reflect physiologically relevant epitopes and binding characteristics
Often polyclonal in nature, recognizing multiple epitopes on the target protein
Can provide insights into disease mechanisms and potential therapeutic approaches
May correlate with disease outcomes (e.g., AD patients with anti-ankyrin G antibodies showed stabilized or improved cognitive scores)
Laboratory-generated monoclonal antibodies (such as A4Mab-1, A4Mab-2, and A4Mab-3 against mACKR4):
Provide consistent, reproducible reagents with defined specificity
Can be precisely characterized for binding affinity (KD values)
Allow targeted epitope selection through immunization strategies
Enable controlled production and quality assessment
Support standardized research applications (e.g., flow cytometry, western blotting)
For receptor proteins like ACR4/ACKR4, laboratory-generated monoclonal antibodies typically offer greater utility for standardized research applications, while natural antibodies may provide valuable insights into disease-relevant immune responses and potential therapeutic approaches. The complementary use of both antibody types could enhance understanding of receptor biology in both normal and pathological contexts.