CAR6 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
CAR6 antibody; EHB1 antibody; At1g70800 antibody; F15H11.5 antibody; F5A18.2 antibody; Protein C2-DOMAIN ABA-RELATED 6 antibody; Protein ENHANCED BENDING 1 antibody
Target Names
CAR6
Uniprot No.

Target Background

Function
This antibody targets Obg-like ATPases, stimulating their GTPase/ATPase activities. It mediates the transient, calcium-dependent interaction of PYR/PYL/RCAR abscisic acid (ABA) receptors with the plasma membrane, thereby regulating ABA sensitivity. Furthermore, it inhibits hypocotyl bending and gravitropic responses under blue light conditions.
Gene References Into Functions
The following studies provide further insight into the target's function: 1. A study of an *Arabidopsis* tropism mutant lacking the EHB1 protein (which interacts with ARF-GAP, and thus indirectly with a small ARF-type G protein) revealed a lower gravitropic threshold for roots and enhanced root bending; however, hypocotyl responses remained largely unaffected. [PMID: 26496692](https://www.ncbi.nlm.nih.gov/pubmed/26496692) 2. EHB1 negatively regulates hypocotyl bending under blue light conditions in *Arabidopsis*, highlighting its role in phototropism. [PMID: 21367967](https://www.ncbi.nlm.nih.gov/pubmed/21367967)
Database Links

KEGG: ath:AT1G70800

STRING: 3702.AT1G70800.1

UniGene: At.35265

Protein Families
Plant CAR protein family
Subcellular Location
Cell membrane. Nucleus.

Q&A

What is CARD6 and what cellular functions does it serve?

CARD6 (Caspase recruitment domain-containing protein 6) is involved in apoptotic signaling pathways and immune regulation. It contains a CARD domain that facilitates protein-protein interactions with other CARD-containing proteins. CARD6 interacts with proteins such as RIP2 in autoimmune disorders like Crohn's disease, and its altered signaling connections can influence cancer progression through effects on cell survival and proliferation mechanisms .

When studying CARD6, researchers should employ multiple methodological approaches to validate findings, including:

  • Protein-protein interaction studies (co-immunoprecipitation, proximity ligation assay)

  • Expression analysis in different tissue types

  • Functional knockdown/knockout experiments to assess phenotypic changes

  • Pathway analysis using specific inhibitors of interacting partners

What experimental applications are suitable for CARD6 antibodies?

CARD6 antibodies can be utilized across multiple experimental platforms. The polyclonal CARD6 antibody (ab227189) has been validated for several applications:

ApplicationValidated StatusRecommended DilutionNotes
Western Blot (WB)Verified1/5000Expected band size: 116 kDa
Immunohistochemistry-Paraffin (IHC-P)Verified1/500Heat-mediated antigen retrieval recommended
Immunocytochemistry/Immunofluorescence (ICC/IF)Verified1/500Compatible with methanol fixation

CARD6 antibodies have been successfully employed to detect expression in human samples including gastric cancer tissue and cell lines such as HeLa (cervical adenocarcinoma) and A549 (lung carcinoma) .

How should CARD6 antibody specificity be validated in research?

Validation of CARD6 antibody specificity requires multiple complementary approaches:

  • Western blot analysis comparing expression across multiple cell lines with known CARD6 expression levels

  • Using positive controls such as A549 cells that demonstrate clear CARD6 expression at the expected molecular weight of 116 kDa

  • Including negative controls through:

    • Preincubation with immunizing peptide

    • Isotype control antibody staining

    • CARD6 knockdown/knockout validation

  • Confirming subcellular localization pattern through ICC/IF matches expected distribution

  • Cross-validating results with alternative CARD6 antibodies from different sources

How can CARD6 antibodies illuminate the protein's role in autoimmune pathways?

CARD6 has significant implications in autoimmune disorders through its interaction with RIP2, which serves as a key mediator in inflammatory signaling cascades. To investigate these interactions:

  • Perform co-immunoprecipitation experiments using CARD6 antibodies in:

    • Healthy tissue samples

    • Inflammatory disease tissues (e.g., Crohn's disease samples)

    • Cell models with inflammatory stimulation (TNF-α, IL-1β)

  • Analyze CARD6-RIP2 complex formation under different conditions:

    • Basal state versus inflammatory stimulus

    • With/without pathway inhibitors

    • Following genetic manipulation of key pathway components

  • Quantify changes in downstream signaling through:

    • Phosphorylation status of NF-κB pathway components

    • Expression of pro-inflammatory cytokines

    • Cellular phenotypic changes

This methodological approach helps elucidate how CARD6 participates in inflammatory signaling networks and potentially identifies intervention points for therapeutic development .

What technical considerations are important when using CARD6 antibodies for cancer research?

When investigating CARD6's role in cancer progression and cell survival mechanisms, researchers should consider:

  • Tissue-specific expression patterns:

    • CARD6 antibodies have been validated in gastric cancer, cervical adenocarcinoma (HeLa), and lung carcinoma (A549) models

    • Expression levels may vary significantly across cancer types and stages

  • Technical optimization for complex cancer tissues:

    • For IHC-P applications, test multiple antigen retrieval methods (heat-mediated versus enzymatic)

    • Optimize antibody concentration (starting with 1/500 dilution for paraffin sections)

    • Consider dual-staining with proliferation or apoptotic markers to correlate with CARD6 expression

  • Functional analysis workflow:

    • Compare CARD6 expression between paired normal/tumor samples

    • Correlate expression with clinical parameters and survival data

    • Perform knockdown/overexpression studies to assess impact on cancer hallmarks (proliferation, apoptosis resistance, migration)

How can CARD6 antibodies be employed to study post-translational modifications?

Studying CARD6 post-translational modifications (PTMs) requires specialized approaches:

  • Immunoprecipitation strategy:

    • Use CARD6 antibodies to pull down the protein from cell lysates

    • Perform mass spectrometry analysis to identify potential PTMs

    • Validate findings with PTM-specific antibodies (phospho, ubiquitin, acetylation, etc.)

  • Differential detection method:

    • Compare total CARD6 detection with modification-specific signals

    • Employ phosphatase or deubiquitinase treatments to confirm specificity

    • Examine changes in PTM patterns following cellular stresses or stimuli

  • Functional correlation:

    • Assess how identified PTMs affect protein-protein interactions

    • Determine if PTMs alter CARD6's subcellular localization

    • Investigate whether PTMs change during disease progression

What are chimeric antigen receptors and how are they constructed?

Chimeric antigen receptors (CARs) are engineered receptor proteins that combine antigen-recognition capabilities with T cell activation domains. Standard CAR structures include:

  • Antigen-binding domain: Typically a single-chain variable fragment (scFv) derived from an antibody that recognizes a specific target antigen

  • Hinge/spacer region: Often derived from IgG4 or CD8α, providing flexibility between binding and signaling domains

  • Transmembrane domain: Usually from CD28 or CD8α, anchoring the CAR in the cell membrane

  • Intracellular signaling domains: Including costimulatory domains (CD28, 4-1BB) and activation domains (CD3ζ)

Second-generation CARs typically contain one costimulatory domain plus CD3ζ, while third-generation CARs incorporate multiple costimulatory domains to enhance T cell activation and persistence .

What methods are used to evaluate CAR expression and functionality?

CAR expression and functionality assessment involves multiple complementary techniques:

  • Expression analysis:

    • Flow cytometry using antibodies against CAR components (e.g., HA-tag staining)

    • Quantification of transduction efficiency in CD4+ and CD8+ T cell populations

    • Assessment of mean fluorescence intensity to determine expression levels

  • Functional assessment:

    • Cytokine secretion assays (IFNγ, TNFα) following target cell exposure

    • Proliferation assays using cell trace violet staining in flow cytometry

    • Cytotoxicity assays against target cells expressing the antigen of interest

  • Specificity testing:

    • Comparison of responses against antigen-positive versus antigen-negative cell lines

    • Dose-response studies with varying antigen densities on target cells

    • Assessment of potential off-target or tonic signaling effects

What are the key differences between CAR generations and their implications for research?

CAR generations differ in their signaling domain architecture, impacting T cell function:

GenerationComponentsAdvantagesLimitationsResearch Applications
1st GenerationscFv + CD3ζSimple designLimited persistence and expansionProof-of-concept studies
2nd GenerationscFv + costimulatory domain (CD28 or 4-1BB) + CD3ζEnhanced expansion and persistencePotential for exhaustionMost current clinical applications
3rd GenerationscFv + multiple costimulatory domains + CD3ζSuperior activation and persistencePotential for increased toxicityAdvanced preclinical models
4th Generation2nd/3rd generation + additional genetic modificationsProgrammable functions (cytokine secretion, etc.)Complex manufacturingCutting-edge research applications

When designing experiments, researchers should select the appropriate CAR generation based on the specific research question, balancing potency with safety considerations .

What approaches exist for developing universal CAR-T cells and what are their experimental advantages?

Universal CAR-T cell platforms represent an advanced research direction with several methodological approaches:

  • Fabrack-CAR system:

    • Utilizes a non-tumor targeted, cyclic meditope peptide as the extracellular domain

    • Binds specifically to engineered binding pockets within monoclonal antibodies (mAbs)

    • Antigen specificity is conferred by administering mAbs with tumor specificity

    • Allows targeting multiple antigens simultaneously by using antibody combinations

  • Split CAR designs:

    • Separates the antigen-recognition and signaling components

    • Enables controlled activation through regulated dimerization

    • Provides improved controllability of CAR effector function

Experimental advantages include:

  • Flexibility to target multiple antigens without manufacturing new CAR-T products

  • Ability to address tumor heterogeneity by using combinations of targeting antibodies

  • Enhanced control over CAR-T activation timing and intensity

  • Facilitation of dose-dependent targeting strategies

  • Potential for reduced manufacturing complexity and cost

These approaches have demonstrated antigen- and antibody-specific T cell activation, proliferation, IFNγ production, selective killing of target cells, and tumor regression in animal models .

How do CAR avidity and epitope selection influence CAR-T cell functionality?

CAR avidity and epitope characteristics significantly impact CAR-T cell performance:

  • Avidity considerations:

    • Higher avidity CARs (lower EC50 values) demonstrate enhanced sensitivity to lower antigen levels

    • The 4D06 and 4D08 CARs showed EC50 values of 0.15 and 0.19 μg/ml, approximately three times higher avidity than the C8-CAR (EC50 of 0.53 μg/ml)

    • Moderate affinity binders may be preferable for the CAR format as they show less off-target toxicity and potentially greater effectiveness

  • Epitope selection factors:

    • Linear versus conformational epitopes affect CAR functionality

    • Linear epitope recognition may contribute to lower tonic signaling

    • Epitope accessibility on the target antigen impacts binding efficiency

    • The binding efficacy of antibodies and their capability to neutralize targets depends on the specific epitope recognized

  • Methodological approach to optimize these parameters:

    • Systematic comparison of CARs with different avidities against the same target

    • Evaluation of tonic signaling through assessment of antigen-independent activation

    • Analysis of CAR-T cell exhaustion markers correlated with avidity measurements

    • Correlation of epitope characteristics with functional outcomes

What experimental strategies can assess and mitigate tonic signaling in CAR-T cells?

Tonic signaling (constitutive activation in the absence of target antigen) presents a critical challenge in CAR-T cell research:

  • Detection methods:

    • Measurement of cytokine production (IFNγ, TNFα) by CAR-T cells exposed to antigen-negative control cells

    • Assessment of CAR-T cell proliferation on antigen-negative targets

    • Analysis of activation markers in the absence of target antigen

  • Experimental findings:

    • 4D08-CAR showed reduced tonic signaling compared to C8-CAR and 4D06-CAR

    • C8-CAR and 4D06-CAR induced IFNγ secretion and pronounced proliferation on HepG2 control cells (antigen-negative)

    • This antigen-independent activation suggests tonic signaling in these constructs

  • Mitigation strategies:

    • Optimization of CAR design (hinge length, transmembrane domain selection)

    • Modification of scFv framework regions to reduce aggregation

    • Tuning of CAR expression levels to minimize baseline activation

    • Exploration of linear epitope-targeting CARs that may exhibit lower tonic signaling

    • Implementation of inducible or split CAR systems for controlled activation

Tonic signaling predisposes CAR-T cells to exhaustion, highlighting the importance of careful construct design and validation to maximize therapeutic potential .

What pre-clinical models best evaluate CAR-T efficacy in solid tumors?

Pre-clinical evaluation of CAR-T cells targeting solid tumors requires robust experimental models:

  • In vitro assessment hierarchy:

    • Target cell line panels with varying antigen expression levels

    • Co-culture systems with effector:target (E:T) ratio optimization

    • 3D tumor spheroid models to better mimic tumor microenvironment

    • Ex vivo patient-derived tumor slices or organoids

  • In vivo modeling approaches:

    • Immunodeficient mouse models (e.g., Bl6.Rag1-/-) for human CAR-T evaluation

    • AAV-delivered viral models to simulate chronic infections (as used for HBV-targeted CARs)

    • Patient-derived xenograft models for heterogeneous tumor representation

    • Humanized mouse models to assess CAR-T function in the context of human immune components

  • Assessment parameters:

    • Measurement of serum viral antigen levels (for viral targets)

    • Monitoring of tissue damage biomarkers (e.g., ALT levels)

    • Quantification of intrahepatic RNA to assess impact on replication

    • Immune histochemistry to evaluate target antigen expression changes

    • Ex vivo restimulation of isolated lymphocytes to assess persistent functionality

How can RNA vaccine approaches enhance CAR-T cell therapy?

CAR-T cell-amplifying RNA vaccine (CARVac) technology represents an innovative approach to enhance CAR-T efficacy:

  • Mechanistic principles:

    • RNA vaccines can be designed to express the target antigen or epitopes

    • This provides in vivo restimulation of CAR-T cells post-infusion

    • The approach enhances CAR-T cell expansion, persistence, and functionality

  • Clinical implementation:

    • Phase 1/2 BNT211-01 trial evaluated CLDN6-specific CAR-T cells with/without CARVac

    • Primary endpoints focused on safety, tolerability, and dose determination

    • Secondary endpoints included objective response rate and disease control rate

  • Experimental outcomes:

    • Manageable toxicity profile with 46% of patients experiencing cytokine release syndrome

    • Only one grade 3 event and 5% with grade 1 immune effector cell-associated neurotoxicity syndrome

    • Encouraging clinical activity observed in patients with relapsed/refractory CLDN6-positive solid tumors

    • Highest response rates in patients with germ cell tumors

This combined approach represents a promising strategy to overcome limitations of conventional CAR-T therapy for solid tumors by providing continued CAR-T stimulation in vivo.

What methodological approaches help identify optimal tumor-associated antigens for CAR-T therapy?

Selection of appropriate tumor-associated antigens requires systematic evaluation:

  • Antigen selection criteria:

    • Expression profile across tumor vs. healthy tissues

    • Surface accessibility for CAR recognition

    • Role in tumor survival/progression (to minimize escape variants)

    • Stability of expression across tumor progression

  • Exemplary target identification:

    • CLDN6 represents an oncofetal cell-surface antigen silenced in healthy adult tissues

    • Expression is strictly suppressed in healthy adult tissues but frequently aberrantly expressed in solid tumors

    • High-level expression detectable in germ cell tumors, epithelial ovarian cancer, endometrial carcinoma and other solid tumors

  • Experimental validation workflow:

    • Prescreening patient samples for target antigen expression

    • Manufacturing CAR-T cells with specificity to the selected antigen

    • Characterizing CAR-T products for phenotype (CD4+/CD8+ composition, memory subsets)

    • Dose escalation studies following a standardized approach (e.g., 3+3 design)

    • Careful monitoring of safety parameters and efficacy outcomes

What are the key challenges in using CAR6 technologies for diverse target applications?

The expansion of CAR technologies faces several methodological challenges that require focused research:

  • Overcoming tumor heterogeneity:

    • Development of universal CAR platforms like Fabrack-CAR that can target multiple antigens through antibody specificity

    • Dual or tandem CAR approaches to minimize antigen escape

    • Exploration of tumor microenvironment targets alongside tumor-specific antigens

  • Improving CAR-T cell persistence and function:

    • Implementation of RNA vaccine approaches to enhance CAR-T expansion in vivo

    • Optimization of CAR signaling domains to balance activation with exhaustion prevention

    • Engineering approaches to overcome immunosuppressive tumor microenvironments

  • Reducing manufacturing complexity:

    • Standardization of CAR-T production protocols

    • Development of allogeneic "off-the-shelf" CAR-T approaches

    • Exploration of non-viral gene transfer methods for CAR introduction

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.