ABCC6 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to ABCC6 Antibody

The ABCC6 protein, encoded by the ABCC6 gene, is a transmembrane transporter primarily expressed in the liver and kidneys . ABCC6 antibodies target this protein for detection in research and diagnostic settings. Their development has been pivotal in studying ABCC6's role in systemic calcification disorders and metabolic pathways .

Applications in Research and Diagnostics

ABCC6 antibodies are utilized in multiple experimental and clinical contexts:

  • Western Blot (WB): Detects ABCC6 protein (~165 kDa) in tissue lysates .

  • Immunohistochemistry (IHC): Localizes ABCC6 in liver and kidney sections, confirming basolateral plasma membrane expression .

  • Immunoprecipitation (IP): Isolates ABCC6 for protein interaction studies .

  • ELISA: Quantifies ABCC6 levels in biological samples .

ABCC6 Localization and Function

  • Basolateral Membrane Localization: Immunohistochemistry using monoclonal antibodies (M6II-7, M6II-24) confirmed ABCC6's presence on the basolateral membrane of hepatocytes, colocalizing with Na,K-ATPase and cadherin .

  • Role in Calcification: ABCC6 deficiency reduces plasma pyrophosphate levels, leading to ectopic calcification in PXE and atherosclerosis . Antibody-based studies in Abcc6−/− mice revealed enlarged vascular calcifications .

Disease-Associated Mutations

  • PXE and GACI: Missense mutations (e.g., R1141X, 23-29del) disrupt ABCC6 transport activity, as shown by immunoblot and immunofluorescence assays .

  • β-Thalassemia: Downregulation of ABCC6 in thalassemic mice was identified using liver-specific antibodies, linking it to secondary calcification .

Therapeutic Insights

  • Pharmacological Rescue: Certain ABCC6 mutants (e.g., R1339C) retained partial transport function in cell models, suggesting potential for targeted therapies .

Technical Considerations and Validation

  • Specificity: Antibodies like D9D1F and K14 are validated using knockout controls (Abcc6−/− mice) to exclude cross-reactivity .

  • Isoforms: ABCC6 has three isoforms; antibodies must be isoform-specific for accurate detection .

  • Tissue Preparation: Frozen sections are preferred for IHC to preserve native protein localization .

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
ABCC6 antibody; MRP6 antibody; MRP8 antibody; At3g13090 antibody; MJG19.4ABC transporter C family member 6 antibody; ABC transporter ABCC.6 antibody; AtABCC6 antibody; EC 7.6.2.2 antibody; ATP-energized glutathione S-conjugate pump 8 antibody; Glutathione S-conjugate-transporting ATPase 8 antibody; Multidrug resistance-associated protein 8 antibody
Target Names
ABCC6
Uniprot No.

Target Background

Function
This antibody targets the glutathione S-conjugate pump.
Database Links

KEGG: ath:AT3G13090

STRING: 3702.AT3G13090.1

UniGene: At.28307

Protein Families
ABC transporter superfamily, ABCC family, Conjugate transporter (TC 3.A.1.208) subfamily
Subcellular Location
Membrane; Multi-pass membrane protein.
Tissue Specificity
Ubiquitous.

Q&A

What is the optimal fixation protocol for immunohistochemical detection of ABCC6 in tissue samples?

For effective immunohistochemical detection of ABCC6 in tissue samples, a sequential fixation protocol is recommended. Begin with 4% paraformaldehyde fixation at 4°C for 4-8 hours, followed by careful washing in phosphate-buffered saline. This approach preserves both the epitope accessibility and membrane architecture where ABCC6 primarily localizes. Since ABCC6 is predominantly expressed in the basolateral plasma membrane of hepatocytes, not in mitochondria-associated membranes as occasionally misreported, membrane integrity preservation is crucial for accurate detection . For frozen liver sections, use acetone fixation for 10 minutes at -20°C to maintain membrane protein epitopes while minimizing background staining. Always include negative controls using samples from ABCC6 knockout mice to validate antibody specificity.

How do I validate the specificity of an ABCC6 antibody for immunoblotting applications?

Validating ABCC6 antibody specificity requires a multi-step approach. First, perform immunoblotting using both positive controls (liver lysates from wild-type models) and negative controls (liver lysates from Abcc6-/- mice) . A specific antibody should detect a protein band at approximately 165 kDa in wild-type samples while showing no reactivity in knockout samples. Second, conduct peptide competition assays by pre-incubating the antibody with the immunizing peptide before immunoblotting. Third, test the antibody on cell lines with confirmed ABCC6 expression versus those lacking expression. Fourth, verify detection of both mature (fully glycosylated) and immature forms of ABCC6, as mutations can affect protein maturation . Finally, perform immunoprecipitation followed by mass spectrometry to confirm the identity of the detected protein.

What epitope regions of ABCC6 are most suitable for antibody generation and why?

The most suitable epitope regions for ABCC6 antibody generation include:

  • N-terminal (aa 1-95) or C-terminal (aa 1427-1503) cytoplasmic domains: These regions are hydrophilic and accessible in both native and denatured states.

  • External loops of transmembrane domains: Particularly regions between TM7-TM8 and TM11-TM12, as these are exposed in the folded protein.

  • Specific regions of nucleotide-binding domains (NBDs): Target unique sequences within NBDs that differ from other ABC transporter family members to prevent cross-reactivity.

Avoid regions with high sequence similarity to other ABCC family transporters, particularly ABCC1 and ABCC3, to prevent cross-reactivity . Additionally, avoid sequences within the Walker A and Walker B motifs of NBD1, as these are highly conserved across the ABC transporter family and may lead to non-specific binding . For detecting structurally compromised ABCC6 variants in PXE research, consider generating antibodies against epitopes that remain accessible even when mutations alter protein folding.

How can I effectively distinguish between different conformational states of ABCC6 using conformation-specific antibodies?

Developing and utilizing conformation-specific antibodies to distinguish between different ABCC6 states requires strategic epitope selection and validation. First, generate antibodies against epitopes that become accessible or hidden during conformational changes associated with the ATP hydrolysis cycle. Target the interface between NBD1 and NBD2, which undergoes significant rearrangement during the transition between ATP-bound and ATP-free states . The NBD1 domain crystallized at 2.3 Å resolution reveals distinct conformational states when bound to nucleotides, providing specific targets for conformation-selective antibodies.

For validation, use ABCC6 protein preparations locked in specific conformations through ATP analogs: ATP-γ-S (non-hydrolyzable) for the ATP-bound state and beryllium fluoride for the post-hydrolysis state. Employ differential antibody binding in native gel electrophoresis, flow cytometry on membrane vesicles, or surface plasmon resonance (SPR) to confirm conformation specificity. Finally, verify functional correlation by comparing antibody binding patterns with transport activity assays to ensure relevance to ABCC6 catalytic cycle.

What strategies can overcome the challenge of detecting low endogenous levels of ABCC6 in non-hepatic tissues?

Detecting low endogenous ABCC6 levels in non-hepatic tissues requires combining enhanced sensitivity techniques with careful controls:

  • Signal amplification methods: Implement tyramide signal amplification (TSA) or rolling circle amplification (RCA) for immunohistochemistry, which can enhance sensitivity by 10-100 fold while maintaining specificity.

  • Proximity ligation assay (PLA): Use two different antibodies targeting distinct ABCC6 epitopes coupled with proximity probes, generating a fluorescent signal only when both epitopes are in close proximity.

  • Ultra-sensitive Western blotting: Employ ECL-Prime or femto-sensitive chemiluminescent substrates combined with tissue concentration techniques such as immunoprecipitation before blotting.

  • Mass spectrometry-based detection: Apply targeted mass spectrometry approaches such as selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) following immunoaffinity enrichment.

For all approaches, include comprehensive controls using tissues from Abcc6-/- models to establish detection thresholds and eliminate false positives . Additionally, validate findings using orthogonal detection methods such as qRT-PCR for transcript levels, acknowledging that transcript and protein levels may not always correlate.

How do different epitope-targeting strategies for ABCC6 antibodies impact detection of disease-causing variants?

Different epitope-targeting strategies significantly impact the detection of ABCC6 disease variants, particularly those causing protein misfolding. Based on structural analysis of ABCC6 NBD1 mutations associated with pseudoxanthoma elasticum (PXE), three distinct antibody approaches should be considered:

  • Conformation-insensitive antibodies: Targeting linear epitopes in regions minimally affected by structural alterations (terminal domains) enables detection of total ABCC6 protein regardless of folding status. These antibodies reveal expression levels but not conformational integrity.

  • Conformation-sensitive antibodies: Targeting epitopes that become inaccessible when mutations disrupt protein folding (particularly regions in the α-helical subdomain) can differentiate between properly folded and misfolded variants. These antibodies have reduced binding to misfolded variants R765Q and A766D compared to wild-type ABCC6 .

  • Domain-specific antibodies: Targeting specific domains like NBD1 or NBD2 can reveal domain-specific folding defects. For instance, antibodies against NBD1 show reduced binding to variants with NBD1 mutations while maintaining reactivity to variants with TMD mutations.

A comprehensive panel approach combining these strategies enables researchers to characterize both the expression level and conformational status of ABCC6 variants. This approach has revealed that approximately 70% of PXE-associated missense mutations cause conformational defects rather than complete absence of protein expression .

What methods effectively differentiate between mature and immature forms of ABCC6 when using antibodies?

Differentiating between mature and immature forms of ABCC6 requires techniques that can distinguish glycosylation states and subcellular localization:

  • Glycosidase digestion analysis: Treat protein samples with Endoglycosidase H (Endo H) and Peptide-N-Glycosidase F (PNGase F) before immunoblotting. Mature (fully processed) ABCC6 is Endo H-resistant but PNGase F-sensitive, while immature forms are sensitive to both enzymes. This approach directly tracks N-glycan processing during protein maturation.

  • Subcellular fractionation: Separate plasma membrane fractions from intracellular organelles (ER/Golgi) using density gradient centrifugation, followed by immunoblotting with ABCC6 antibodies. Mature ABCC6 predominantly localizes to the plasma membrane fraction.

  • Pulse-chase analysis: Metabolically label cells with 35S-methionine/cysteine, then immunoprecipitate ABCC6 at various chase time points. The mature form appears later in the chase period and shows a higher molecular weight due to complex glycosylation.

  • Deglycosylation mobility shift assay: Compare the electrophoretic mobility of ABCC6 in samples before and after complete deglycosylation. The difference in molecular weight between native and deglycosylated forms is greater for mature ABCC6 (with complex glycans) than for immature forms (with high-mannose glycans).

These methods are particularly valuable when studying PXE-associated mutations that affect ABCC6 processing, as many variants show defective maturation despite normal initial expression .

How can I optimize immunoprecipitation protocols for ABCC6 given its membrane protein characteristics?

Optimizing immunoprecipitation (IP) of ABCC6 requires protocols specifically designed for hydrophobic membrane proteins:

  • Membrane solubilization: Use mild detergents like 1% digitonin, 0.5-1% n-dodecyl-β-D-maltoside (DDM), or 1% CHAPS that preserve native protein conformation while effectively solubilizing membrane-embedded ABCC6. Avoid harsh detergents like SDS that denature epitopes.

  • Buffer optimization: Include 10% glycerol, 5 mM MgATP, and 150 mM NaCl in all buffers to stabilize ABCC6 structure during extraction. ATP binding significantly enhances ABCC6 stability, particularly its NBD domains .

  • Cross-linking approach: For transient or weak protein-protein interactions, incorporate a membrane-permeable cross-linker (DSP or formaldehyde at 0.5-1%) prior to cell lysis.

  • Antibody selection: Use antibodies targeting extracellular loops or cytoplasmic domains rather than transmembrane regions, which are often inaccessible in the solubilized state.

  • Sequential immunoprecipitation: For studying ABCC6 complexes, perform sequential IPs with antibodies against suspected interaction partners, followed by ABCC6 antibodies to verify specific associations.

This optimized protocol has successfully isolated both wild-type ABCC6 and disease-associated variants, enabling comparative proteomic analysis of their interaction networks. For challenging ABCC6 variants with altered conformation, increasing the detergent-to-protein ratio and extending incubation times may improve solubilization efficiency.

What controls are essential when using ABCC6 antibodies to study protein localization in disease models?

When investigating ABCC6 localization in disease models, implement these essential controls:

  • Genetic negative controls: Include tissues/cells from Abcc6 knockout models processed identically to experimental samples. This definitively establishes antibody specificity and background signal levels .

  • Peptide competition controls: Pre-incubate ABCC6 antibody with excess immunizing peptide before application to verify signal specificity.

  • Multiple antibody validation: Use at least two antibodies targeting different ABCC6 epitopes to confirm localization patterns. Consistent results across different antibodies strongly support accurate detection.

  • Subcellular marker co-localization: Co-stain with established markers for plasma membrane (Na+/K+-ATPase), ER (calnexin), Golgi (GM130), and mitochondria (TOMM20) to precisely define ABCC6 localization. This is critical given past controversies regarding ABCC6 localization to mitochondria-associated membranes versus plasma membrane .

  • Transfected controls: Include cells expressing tagged ABCC6 constructs (GFP or FLAG) to compare antibody-based detection with tag-based detection.

  • Cross-reactivity assessment: Test ABCC6 antibodies on tissues from other species or on cells expressing closely related transporters (ABCC1, ABCC3) to ensure specificity within the ABC transporter family.

These controls are particularly important when studying disease-associated ABCC6 variants that may exhibit altered trafficking patterns, such as the retention in ER or Golgi observed in certain PXE-associated mutations .

How should researchers interpret discrepancies between ABCC6 protein levels detected by different antibodies?

When faced with discrepancies between ABCC6 protein levels detected by different antibodies, researchers should implement a systematic analysis approach:

  • Epitope accessibility analysis: Map the epitopes of each antibody to the ABCC6 structure. Antibodies targeting different domains (NBDs versus TMDs) may show discrepant results if mutations or conditions alter domain-specific folding. The high-resolution structure of NBD1 at 2.3 Å provides a template for understanding epitope accessibility in this domain .

  • Conformational sensitivity assessment: Test antibodies against ABCC6 under different conditions that alter protein conformation (varying ATP concentrations, detergent types, temperature). Some antibodies may preferentially recognize specific conformational states of ABCC6.

  • Glycosylation interference evaluation: Determine if glycosylation masks epitopes by comparing antibody binding before and after deglycosylation. ABCC6 undergoes extensive glycosylation during maturation, which can shield certain epitopes.

  • Cross-reactivity verification: Perform immunoprecipitation followed by mass spectrometry to identify all proteins recognized by each antibody, verifying whether discrepancies result from off-target binding to other ABC transporters.

  • Quantitative calibration: Develop a standard curve using purified recombinant ABCC6 domains to calibrate each antibody's detection efficiency and linear range.

When reporting discrepant findings, present data from multiple antibodies alongside these validation analyses to enable accurate interpretation of ABCC6 expression levels, particularly important when studying disease-associated variants with altered conformation .

What methodological approaches can reconcile contradictory findings regarding ABCC6 subcellular localization?

Contradictory findings regarding ABCC6 subcellular localization, particularly the debate between plasma membrane versus mitochondria-associated membrane localization, can be reconciled through these methodological approaches:

  • Multiscale imaging analysis: Combine confocal microscopy with super-resolution techniques (STORM, PALM) to achieve nanoscale resolution beyond the diffraction limit. This distinguishes between true co-localization and mere proximity of subcellular structures.

  • Complementary biochemical fractionation: Perform density gradient fractionation with differential centrifugation to physically separate subcellular compartments, followed by immunoblotting for ABCC6 and compartment-specific markers.

  • Functional location verification: Implement surface biotinylation assays that selectively label plasma membrane proteins, distinguishing surface-exposed ABCC6 from intracellular pools.

  • Electron microscopy with immunogold labeling: Utilize this technique for the highest resolution localization, capable of precisely distinguishing between plasma membrane and closely associated membrane structures.

  • Live-cell tracking with split fluorescent proteins: Use complementation systems where one fragment is targeted to specific organelles while the other is fused to ABCC6, generating fluorescence only when ABCC6 enters specific compartments.

These approaches have conclusively demonstrated that ABCC6 primarily localizes to the basolateral plasma membrane of hepatocytes rather than mitochondria-associated membranes as previously suggested in some studies . This correct localization is consistent with ABCC6's proposed function in cellular transport processes and its role in PXE pathophysiology.

How can researchers distinguish between primary effects of ABCC6 deficiency and secondary compensatory responses in disease models?

Distinguishing primary ABCC6 deficiency effects from secondary compensatory responses requires temporal and mechanistic dissection strategies:

  • Time-course analyses: Implement longitudinal studies beginning before phenotype manifestation. In ABCC6-deficient models, collect samples at multiple time points from early development through disease progression to identify temporal sequence of molecular changes.

  • Inducible knockout systems: Utilize tetracycline-regulated or tamoxifen-inducible Cre-loxP systems to induce ABCC6 deficiency at defined time points, enabling distinction between immediate (primary) responses and later (secondary) adaptations.

  • Transcriptomic and proteomic comparisons: Compare acute (24-72h) versus chronic (weeks-months) gene and protein expression profiles following ABCC6 loss. Primary effects typically show immediate alterations that persist, while compensatory responses emerge gradually.

  • Pathway-focused phosphoproteomics: Assess cellular signaling pathway activation states through quantitative phosphoproteomic analysis at multiple time points following ABCC6 loss to distinguish initial signaling disruptions from feedback responses.

  • Cross-species comparative analysis: Compare molecular signatures across different ABCC6-deficient models (mouse, zebrafish, cell lines) to identify conserved primary responses versus species-specific secondary adaptations.

  • Rescue experiments with temporal control: Reintroduce ABCC6 at different disease stages to determine which phenotypes are reversible (likely primary) versus established (potentially secondary).

These approaches have helped clarify that ectopic mineralization in pseudoxanthoma elasticum results primarily from ABCC6 deficiency rather than from secondary inflammatory responses or compensatory calcium metabolism changes .

How can ABCC6 antibodies be utilized to develop therapeutic monitoring assays for emerging PXE treatments?

ABCC6 antibodies can be strategically employed to develop therapeutic monitoring assays for emerging PXE treatments through these methodological approaches:

  • Conformation-specific biomarker assays: Develop sandwich ELISA systems using capture antibodies against invariant ABCC6 epitopes and detection antibodies against conformation-dependent epitopes. This configuration enables quantification of properly folded ABCC6 protein in patient samples during treatment with molecular chaperones or corrector therapies.

  • Trafficking assessment systems: Implement cell-based assays using polarized hepatic cell lines expressing disease-associated ABCC6 variants. Apply antibodies targeting extracellular epitopes in non-permeabilized cells to quantify surface expression following treatment with trafficking correctors.

  • Functional recovery monitoring: Combine ABCC6 antibody-based detection with functional transport assays using known or candidate ABCC6 substrates. This correlation between protein expression and function provides comprehensive therapeutic efficacy assessment.

  • Tissue-specific correction measurement: Develop immunohistochemical scoring systems based on ABCC6 antibody staining patterns in skin biopsies from PXE patients undergoing treatment, establishing a quantitative relationship between hepatic ABCC6 correction and amelioration of peripheral tissue calcification.

These monitoring approaches are particularly valuable for emerging therapeutic strategies targeting ABCC6 folding and trafficking defects, which constitute approximately 70% of PXE-causing mutations . The methods provide quantitative metrics beyond clinical assessment to evaluate treatment efficacy at the molecular level.

What protocols effectively apply ABCC6 antibodies to study protein-protein interactions that influence transporter function?

Studying ABCC6 protein-protein interactions requires specialized protocols optimized for membrane protein complexes:

  • Membrane-based co-immunoprecipitation: Solubilize membranes using digitonin (0.5-1%) or DDM (0.1-0.5%) to preserve native protein complexes. Perform immunoprecipitation with ABCC6 antibodies followed by immunoblotting for suspected interaction partners or mass spectrometry for unbiased discovery.

  • Proximity labeling proteomics: Express ABCC6 fused to promiscuous biotin ligases (BioID2 or TurboID) in relevant cell types. The biotin ligase biotinylates proteins in close proximity to ABCC6, which are then purified with streptavidin and identified by mass spectrometry.

  • FRET/BRET-based interaction assays: Develop live-cell assays using ABCC6 fused to donor fluorophores and candidate interactors fused to acceptor fluorophores. Energy transfer occurs only when proteins interact, providing both spatial and temporal interaction information.

  • Split-luciferase complementation: Express ABCC6 and potential interaction partners as fusions with complementary luciferase fragments, generating luminescence only upon protein interaction.

  • Chemical cross-linking mass spectrometry (XL-MS): Apply membrane-permeable crosslinkers to stabilize transient interactions, followed by ABCC6 immunoprecipitation, enzymatic digestion, and mass spectrometry to identify crosslinked peptides that reveal interaction interfaces.

These approaches have identified interactions between ABCC6 and other membrane proteins involved in calcification regulation, chaperones that facilitate folding, and cytoskeletal components that influence membrane localization. Understanding these interactions provides insight into both normal ABCC6 function and disease mechanisms in PXE .

How can researchers apply ABCC6 antibodies to validate gene therapy approaches for ABCC6-related disorders?

Validating gene therapy approaches for ABCC6-related disorders requires comprehensive antibody-based assessment protocols:

  • Multi-level expression analysis: Implement a hierarchical detection strategy using ABCC6 antibodies to verify:

    • Transgene expression at the protein level (total ABCC6 content)

    • Proper protein processing (ratio of mature to immature forms)

    • Correct subcellular localization (basolateral plasma membrane targeting)

    • Tissue-specific expression patterns (primarily liver with minor expression in kidneys)

  • Epitope-specific detection for vector-derived protein: Develop assays that specifically detect vector-encoded ABCC6 versus endogenous protein:

    • For epitope-tagged constructs: Use antibodies against the tag in parallel with ABCC6 antibodies

    • For untagged constructs: Develop allele-specific antibodies that distinguish therapeutic from mutant versions through single amino acid differences

    • For species-specific constructs: Use antibodies that discriminate between human therapeutic ABCC6 and endogenous mouse Abcc6 in preclinical models

  • Functional correction assessment: Correlate ABCC6 protein restoration with:

    • Normalization of pyrophosphate metabolism

    • Reduction in ectopic calcification measured by histological analysis

    • Improvement in tissue elasticity measurements

This comprehensive antibody-based validation strategy enables researchers to determine whether gene therapy achieves physiologically relevant ABCC6 expression in appropriate cellular compartments with functional outcomes. The approach has successfully validated AAV-based gene therapy approaches in Abcc6-/- mouse models, demonstrating both hepatic expression of functional ABCC6 and reduction in peripheral tissue calcification .

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.