ATF6 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
Orders are typically dispatched within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines.
Synonyms
ATF6; Cyclic AMP-dependent transcription factor ATF-6 alpha; cAMP-dependent transcription factor ATF-6 alpha; Activating transcription factor 6 alpha; ATF6-alpha
Target Names
Uniprot No.

Target Background

Function

This antibody targets the precursor form of the activating transcription factor 6 alpha (ATF6α), a transcription factor residing within the endoplasmic reticulum (ER) membrane. Under ER stress conditions, this precursor undergoes processing, releasing the active transcription factor which translocates to the nucleus. Here, it activates transcription of genes involved in the unfolded protein response (UPR), a cellular mechanism mitigating ER stress. ATF6α binds to specific DNA sequences, including the ER stress response element (ERSE) and ERSE II, often in conjunction with NF-Y. Beyond its role in the UPR, ATF6α may also regulate transcription via the serum response factor and potentially contributes to foveal development and cone function in the retina.

Gene References Into Functions

The following studies highlight the diverse roles of ATF6α:

  1. Compound 147 preferentially activates ATF6 signaling through localized metabolic activation and selective modification of ER proteins regulating ATF6 activity. PMID: 30084354
  2. Sustained intestinal ATF6 activation promotes colonic dysbiosis and microbiota-dependent tumorigenesis. PMID: 30063920
  3. ATF6α plays a crucial role in establishing and maintaining cellular senescence in normal human fibroblasts by upregulating a COX2/PGE2 intracrine pathway. PMID: 28803844
  4. High activated ATF6α expression exacerbates ER stress-induced vascular endothelial cell apoptosis via the mitochondrial apoptotic pathway, upregulating caspase-3, caspase-9, CHOP, cytochrome c, and Bax/Bcl2. PMID: 29512699
  5. Phenobarbital-induced ER membrane expansion depends on ATF6 transmembrane domain-induced effects. PMID: 30086303
  6. ATF6 plays a distinct role in viral protein stability, with the host employing unconventional cleavage mechanisms to combat viral infection. PMID: 29386036
  7. ATF6 is one of three branches of the UPR, alongside IRE1 and PERK. PMID: 28105371
  8. ASNS expression is significantly elevated with ATF6 overexpression; both are decreased in hepatocellular carcinoma (HCC), with a positive correlation between their mRNA levels. rs34050735 is associated with HCC and overall survival. PMID: 28629319
  9. Human ATF6 mutations disrupt sequential steps in ATF6 activation. PMID: 28028229
  10. ATF6α is central to the establishment of senescence morphology in primary fibroblasts. PMID: 27563820
  11. A novel homozygous ATF6 mutation (c.1691A>G, p.(Asp564Gly)) is associated with autosomal recessive cone-rod dystrophy. PMID: 28812650
  12. Genetic mutations in ATF6 can cause achromatopsia. PMID: 27117871
  13. ATF6 is negatively regulated through post-translational SUMOylation. PMID: 29061306
  14. Low ATF6 expression is associated with cancer. PMID: 28157699
  15. The ATF6α pathway and ER-associated protein degradation (ERAD) are elevated in salivary glands of Sjögren's syndrome patients. PMID: 27461470
  16. An ATF6/C/EBP-β transcription factor complex is required for IFN-γ-induced DAPK1 expression. PMID: 27590344
  17. Studies examining ATF6 cleavage, MRLC phosphorylation, DAPK1 expression, DAPK1 transcription, and the subcellular localization of ATF6 and mAtg9. PMID: 27085326
  18. Genetic variation in ATF6 (rs2070150) is not associated with HCC in Thai patients with HBV infection. PMID: 26925648
  19. High-content screening identified compounds that inhibit ATF6 nuclear translocation. PMID: 26707144
  20. DREAM silencing activates ATF6 signaling, promoting neuroprotection in Huntington's disease. PMID: 26752648
  21. ATF6 and caspase-12 trigger apoptosis in neonatal hypoxic-ischemic encephalopathy. PMID: 26261584
  22. HIV infection activates stress-response components, and antiretroviral therapy alters UPR activation. PMID: 25976933
  23. Defective podocyte insulin signaling promotes ATF6-dependent maladaptive ER stress in diabetic nephropathy. PMID: 25754093
  24. Increased protein expression of ER stress markers in placentas of women with preeclampsia. PMID: 25675914
  25. Autosomal recessive achromatopsia caused by a frameshift mutation in ATF6. PMID: 26063662
  26. ATF6A plays a crucial role in foveal development and cone function. PMID: 26029869
  27. ATF6 reduces secretion and extracellular aggregation of destabilized TTR variants. PMID: 25444553
  28. ATF6 genetic variation is associated with prediabetes and interacts with BMI. PMID: 25302688
  29. Silver nanoparticles induce ATF6 degradation, activating the NLRP-3 inflammasome and pyroptosis. PMID: 25593314
  30. CiC expression is activated during ER stress through ATF6α and XBP1 binding to a UPRE element. PMID: 25450523
  31. Mutation of p38 MAPK phosphorylation sites in ATF6 suppresses DAPK1 induction. PMID: 25135476
  32. ATF6 single nucleotide polymorphism is associated with increased BMI after kidney transplantation. PMID: 22099811
  33. ATF6 and PERK signaling protect melanoma cells from ER stress-induced apoptosis. PMID: 24240056
  34. ATF6 binds to the XBP1 promoter and enhances XBP1S expression in osteoarthritis cartilage. PMID: 24269637
  35. ATF6 pathway activation limits ATZ-dependent cell toxicity by promoting ERAD of ATZ. PMID: 21976666
  36. Activation of IRE1, ATF6, or PERK prevents mutant rhodopsin accumulation in cells. PMID: 24664756
  37. Tick-borne encephalitis virus infection activates the IRE1 pathway and ATF6. PMID: 24177270
  38. PolyP and isoquercitrin co-enhance bone mineral formation, potentially useful for osteoporosis treatment. PMID: 24726443
  39. ATF6 mRNA expression decreases with disease progression. PMID: 24302549
  40. ATF6α regulates de novo cholesterol synthesis by stimulating cholesterogenic gene expression. PMID: 23924739
  41. Mediator subunit MED25 plays a critical role in ATF6α activation. PMID: 23864652
  42. ATF6 is an ERAD-Lm substrate requiring SEL1L for degradation. PMID: 24043630
  43. ATF6 and CHOP are involved in ox-LDL-induced cholesterol accumulation and apoptosis in macrophages. PMID: 23037953
  44. ATF6 activation is significantly higher for the CASPR2-D1129H protein compared to wild-type. PMID: 22872700
  45. ATF6 is one of three ER transmembrane protein sensors signaling the UPR. PMID: 22917505
  46. ORMDL3 transfection induced expression of several genes and selectively activated ATF6. PMID: 23011799
  47. Selective activation of ATF6 or PERK prevents mutant rhodopsin accumulation. PMID: 22956602
  48. ATF6α recruits RNA polymerase II coregulatory complexes to ER stress response enhancer elements. PMID: 22577136
  49. Increased expression of GRP78, ATF6, and CHOP in endometrioid endometrial carcinomas suggests a role for ER stress in endometrial cancer. PMID: 22146569
  50. BMP2 induces osteoblast differentiation through Runx2-dependent ATF6 expression, regulating Oc transcription. PMID: 22102412
Database Links

HGNC: 791

OMIM: 605537

KEGG: hsa:22926

STRING: 9606.ENSP00000356919

UniGene: Hs.492740

Involvement In Disease
Achromatopsia 7 (ACHM7)
Protein Families
BZIP family, ATF subfamily
Subcellular Location
Endoplasmic reticulum membrane; Single-pass type II membrane protein. Golgi apparatus membrane; Single-pass type II membrane protein.; [Processed cyclic AMP-dependent transcription factor ATF-6 alpha]: Nucleus.
Tissue Specificity
Ubiquitous.

Q&A

What is ATF6 and why is it important in cellular research?

ATF6 (Activating Transcription Factor 6) is a key transcription factor involved in the unfolded protein response (UPR), which is activated during endoplasmic reticulum (ER) stress. Research has demonstrated that ATF6 plays a critical role in regulating CHOP (C/EBP homologous protein) dynamics, which in turn influences cell fate decisions during stress responses . The protein exists in different forms - primarily as a 90 kDa full-length transmembrane protein in unstressed conditions and as cleaved forms (ranging from 36-70 kDa) that translocate to the nucleus under stress conditions . ATF6 research is particularly important because it represents one of the three main branches of the UPR (alongside IRE1 and PERK pathways), and mathematical modeling has confirmed that ATF6 is essential for properly describing the dynamic peak in CHOP expression during cellular stress responses .

What are the different forms of ATF6 and how can they be detected?

ATF6 exists in multiple forms that reflect its activation status and cellular localization:

  • Full-length ATF6: The primary form is approximately 90-100 kDa and is typically anchored in the ER membrane in unstressed cells .

  • Cleaved/active forms: Under ER stress, ATF6 undergoes proteolytic cleavage, generating multiple active fragments:

    • 50-70 kDa cleaved forms observed following treatment with stressors like azetidine (AzC)

    • A 50 kDa nuclear form particularly prominent in certain tumor cell lines

    • 60 kDa and 36 kDa forms detected in the nucleus

Detection methods vary by application, with Western blotting commonly used to distinguish between full-length and cleaved forms. Immunofluorescence allows visualization of subcellular localization, with ATF6 typically observed in the ER under normal conditions and in the nucleus during stress . Flow cytometry can be used to quantify ATF6 expression at the cellular level, requiring permeabilization for access to intracellular ATF6 .

What are the standard applications for ATF6 antibodies in research?

ATF6 antibodies are utilized across multiple experimental applications, each providing different insights into protein expression, localization, and function:

ApplicationDescriptionTypical DilutionKey Considerations
Western Blot (WB)Detection of full-length and cleaved ATF6 forms1:5000-1:50000 May require sensitive ECL substrates
Immunohistochemistry (IHC)Visualization in tissue sections1:250-1:1000 Epitope retrieval methods impact results
Immunofluorescence (IF)/ICCSubcellular localization studies1:400-1:1600 Methanol fixation at -20°C recommended
Flow Cytometry (FC)Quantitative analysis at cellular level0.40 μg per 10^6 cells Requires fixation and permeabilization
Chromatin Immunoprecipitation (ChIP)Study of DNA-protein interactions1:10-1:500 Useful for identifying ATF6 binding sites
ImmunoprecipitationProtein complex isolationApplication-specificUseful for studying protein interactions

These applications have been validated across multiple cell types including HeLa, HEK-293, MCF-7, and various cancer cell lines, making ATF6 antibodies versatile tools for both basic and advanced research questions .

How can researchers differentiate between ATF6α and ATF6β in experimental systems?

Distinguishing between ATF6α and ATF6β isoforms represents a significant challenge in research due to their structural similarities. Available evidence indicates that some ATF6 antibodies, such as the monoclonal antibody described in the search results (70B1413.1), are specific for ATF6α and do not recognize ATF6β . This specificity was confirmed in research by Bommiasamy et al. (2009), providing a valuable tool for researchers needing to distinguish between these isoforms .

For experimental design, researchers should consider:

  • Antibody selection: Verify isoform specificity in the antibody documentation before designing experiments.

  • Control experiments: Include ATF6α and ATF6β overexpression controls to confirm antibody specificity.

  • Western blot analysis: ATF6α typically appears at 90-100 kDa while cleaved forms show distinct banding patterns.

  • Genetic approaches: Complement antibody-based detection with siRNA knockdown of specific isoforms to confirm specificity.

  • Functional assays: Since ATF6α and ATF6β have distinct roles in the UPR, functional readouts (such as downstream target activation) can help confirm isoform identity.

When publishing results, researchers should clearly specify which isoform was studied and provide validation data confirming antibody specificity to avoid confusion in the literature .

What methodological approaches best capture the dynamic changes in ATF6 localization during ER stress responses?

Capturing the dynamic translocation of ATF6 from the ER to the Golgi and subsequently to the nucleus during stress responses requires sophisticated methodological approaches:

  • Live-cell imaging: For real-time visualization of ATF6 translocation, researchers can use:

    • Fluorescently-tagged ATF6 constructs

    • Time-lapse microscopy following stress induction

    • Co-localization studies with ER, Golgi, and nuclear markers

  • Subcellular fractionation: To quantify ATF6 distribution across cellular compartments:

    • Separate nuclear, cytoplasmic, and membrane fractions

    • Perform Western blotting with ATF6 antibodies on each fraction

    • Normalize to compartment-specific markers (lamin for nucleus, calnexin for ER)

  • Immunofluorescence microscopy: Fixed-cell approaches should:

    • Use methanol fixation at -20°C for optimal ATF6 detection

    • Include time-course experiments (15min, 30min, 1h, 2h, 4h post-stress)

    • Perform co-staining with organelle markers

    • Use confocal microscopy for precise localization

  • Flow cytometry: For population-level quantification:

    • Apply the recommended fixation and permeabilization methods using FlowX FoxP3 Fixation & Permeabilization Buffer Kit

    • Include time-course measurements following stress induction

    • Consider dual staining for full-length and cleaved forms

  • ChIP assays: To confirm nuclear translocation and activity:

    • Perform chromatin immunoprecipitation at different time points following stress

    • Analyze binding to known ATF6 target sequences

The search results indicate that untreated HeLa cells show predominantly ER localization of ATF6, which can serve as an important baseline control for stress-induced translocation experiments .

How does ATF6 activation impact CHOP dynamics and cell fate decisions in different model systems?

Mathematical modeling and experimental evidence indicate that ATF6 plays a crucial role in shaping CHOP dynamics, particularly in the early phases of the unfolded protein response . This relationship has significant implications for cell fate decisions in various model systems:

  • Mathematical modeling evidence:

    • Models incorporating ATF6 accurately describe the dynamic peak in CHOP expression observed experimentally

    • ATF6-free models fail to capture this peak, yielding statistically inferior fits (ΔG = 119 vs. 271)

    • Information criteria analyses (AIC and BIC) strongly favor models including the ATF6 branch (AIC: 332 vs. 620)

  • Experimental approaches to study this relationship:

    • Time-course experiments measuring both ATF6 cleavage and CHOP expression

    • ChIP assays to confirm ATF6 binding to CHOP promoter elements

    • ATF6 knockdown/knockout systems to assess impact on CHOP dynamics

    • Single-cell analyses to capture cell-to-cell variability in responses

  • Cell type-specific responses:

    • Certain tumor cell lines show constitutive expression of the cleaved 50 kDa nuclear form of ATF6, including:

      • B cell lymphoma (DEL)

      • Primary effusion lymphoma [BC-3, PEL-SY, HBL-6]

      • Lymphoblastic leukemia (DS-1)

      • Multiple myeloma lines (RPMI-8226, NCI-H929)

    • These constitutive expression patterns may contribute to altered stress responses and cell survival mechanisms in malignant cells

  • Potential methodological approaches:

    • Reporter systems for real-time monitoring of CHOP expression

    • Dual-color systems to simultaneously track ATF6 and CHOP

    • CRISPR-mediated tagging of endogenous proteins to maintain physiological expression levels

    • Pharmacological inhibitors of ATF6 processing to assess temporal requirements

Understanding this relationship provides important insights into how cells determine whether to adapt to stress or initiate apoptosis, with significant implications for diseases involving ER stress, including neurodegenerative conditions, diabetes, and cancer .

What are the optimal conditions for detecting cleaved forms of ATF6 by Western blotting?

Detecting cleaved forms of ATF6 by Western blotting presents technical challenges that require specific optimization strategies:

  • Sample preparation considerations:

    • Induce ER stress to generate cleaved forms (e.g., using azetidine, tunicamycin, thapsigargin)

    • Include protease inhibitors in lysis buffers to prevent artificial degradation

    • Consider preparing nuclear extracts to enrich for cleaved forms

    • Use fresh samples when possible, as freeze-thaw cycles may affect detection

  • Technical optimization parameters:

    • Gel percentage: 8-10% gels typically provide better resolution in the 36-100 kDa range

    • Transfer conditions: Longer transfer times (overnight at low voltage) may improve transfer of larger proteins

    • Blocking: Use 5% non-fat dry milk or BSA depending on antibody specifications

    • Primary antibody dilution: Follow recommended dilutions (1:5000-1:50000) , but may require optimization

    • Detection system: Use maximum sensitivity ECL substrate (Femto sensitive) for optimal detection

  • Controls to include:

    • Positive control: The ATF6 transfected cell lysate is recommended as a useful western blot positive control

    • Stress conditions: Include both stressed and unstressed samples

    • Size markers: Ensure markers cover the 36-100 kDa range to identify all forms

    • Loading controls: Use appropriate controls for whole cell lysates, nuclear fractions, or membrane fractions

  • Expected banding patterns:

    • Full-length ATF6: 90-100 kDa band

    • Cleaved forms: Multiple bands may appear between 50-70 kDa following stress induction

    • Additional cleaved forms: 60 kDa and 36 kDa forms have been described

    • Note that banding patterns may vary between cell types and stress conditions

This methodological approach helps researchers reliably detect and distinguish between full-length and cleaved forms of ATF6, enabling more accurate assessment of UPR activation in experimental systems.

How should researchers optimize immunofluorescence protocols for ATF6 detection in different cell types?

Optimizing immunofluorescence protocols for ATF6 detection requires careful consideration of fixation, permeabilization, and antibody incubation conditions:

  • Fixation and permeabilization:

    • Methanol fixation at -20°C for 10 minutes is recommended based on published protocols

    • Alternative fixation methods (4% paraformaldehyde) may be tested if methanol proves problematic

    • For paraformaldehyde fixation, additional permeabilization with 0.1-0.5% Triton X-100 is typically required

    • Air-dry fixed cells briefly before rehydration in PBS for 5 minutes at room temperature

  • Antibody selection and dilution:

    • For immunofluorescence/ICC, recommended dilution ranges from 1:400-1:1600 or 1:10-1:500 depending on the antibody

    • Consider using monoclonal antibodies for consistent results across experiments

    • Select antibodies validated for immunofluorescence applications in your cell type of interest

  • Protocol optimization for different cell types:

    • Adherent epithelial cells (e.g., HeLa, A549): Standard protocols work well

    • Suspension cells: Consider cytospin preparation or attachment to poly-L-lysine coated slides

    • Primary cells: May require gentler fixation conditions and higher antibody concentrations

    • Tissue sections: Additional optimization of antigen retrieval methods may be necessary

  • Co-staining considerations:

    • For subcellular localization studies, consider co-staining with markers for:

      • ER (e.g., calnexin, PDI)

      • Golgi (e.g., GM130)

      • Nucleus (e.g., DAPI)

    • Select secondary antibodies with non-overlapping emission spectra

    • Include appropriate controls for each fluorophore

  • Visualization parameters:

    • Confocal microscopy provides superior resolution for subcellular localization

    • Use consistent exposure settings when comparing conditions

    • For stress response studies, include a time course (e.g., 0, 1, 3, 6, 12 hours)

Example protocol from the search results: Untreated HeLa cells were fixed in -20°C methanol for 10 min, air dried and rehydrated in PBS at room temperature for 5 minutes. Cells were incubated with anti-ATF6 (1:20) for one hour at room temperature. ATF6 reactivity was detected with anti-mouse Dylight-488 secondary antibody. Nuclei were counterstained with DAPI. This approach successfully demonstrated ER localization of ATF6 in unstressed conditions .

What controls and validation steps are essential when using ATF6 antibodies in research?

Rigorous controls and validation steps are critical for ensuring the reliability and reproducibility of ATF6 antibody-based experiments:

  • Essential controls for antibody validation:

    • Positive controls: Use cell lines known to express ATF6 (e.g., HeLa, U2OS, HEK-293)

    • Negative controls: Include secondary antibody-only controls and isotype controls (e.g., rabbit IgG control antibody)

    • ATF6 overexpression: Transfected cell lysates can serve as positive controls

    • Knockout/knockdown controls: ATF6 knockout or knockdown cells provide critical specificity validation

  • Validation across different applications:

    • Western blot: Confirm expected molecular weight (90-100 kDa for full-length, 36-70 kDa for cleaved forms)

    • Immunofluorescence: Verify expected subcellular localization (ER in unstressed cells, nuclear during stress)

    • Flow cytometry: Compare with isotype control to establish specific staining

    • IHC: Include positive and negative tissue controls with established staining patterns

  • Stress induction validation:

    • Treatment with ER stressors should induce predictable changes in ATF6 processing

    • Azetidine (AzC) treatment has been documented to induce cleaved forms in the 50-70 kDa range

    • Time-course experiments can validate the expected temporal dynamics of ATF6 processing

  • Cross-reactivity assessment:

    • Confirm isoform specificity (ATF6α vs. ATF6β)

    • Test for cross-reactivity with related ATF family members

    • If working across species, validate species-specific reactivity

  • Technical validation parameters:

    • Antibody dilution optimization: Titrate antibodies to determine optimal working concentration

    • Epitope retrieval methods: For IHC, compare different retrieval methods (e.g., TE buffer pH 9.0 vs. citrate buffer pH 6.0)

    • Reproducibility assessment: Confirm consistent results across different lots of the same antibody

  • Application-specific validation:

    • For ChIP applications: Validate enrichment of known ATF6 target genes

    • For co-IP experiments: Confirm interaction with known binding partners

    • For flow cytometry: Optimize fixation and permeabilization conditions using the recommended buffer systems

Implementing these validation steps ensures that experimental findings are truly reflective of ATF6 biology rather than artifacts of antibody cross-reactivity or technical limitations.

Why might researchers observe multiple banding patterns when detecting ATF6 by Western blotting?

Multiple banding patterns in ATF6 Western blots are common and can reflect both biological and technical factors:

  • Biological factors contributing to multiple bands:

    • Different ATF6 forms: Full-length (90-100 kDa) and various cleaved forms (36-70 kDa)

    • Stress conditions: Treatment with ER stressors induces proteolytic processing, generating additional bands

    • Cell type specificity: Different cell lines may show variations in processing patterns

    • Post-translational modifications: Glycosylation and phosphorylation can alter mobility

    • Isoforms: ATF6α and ATF6β may be detected differently depending on antibody specificity

  • Technical factors that may generate multiple bands:

    • Partial degradation during sample preparation

    • Incomplete reduction of disulfide bonds

    • Non-specific antibody binding

    • Sample overloading causing band distortion

    • Insufficient blocking leading to background bands

  • Published observations on banding patterns:

    • In landmark ATF6 studies, multiple bands were observed between 66-116 kDa and 45-66 kDa in both unstressed and stressed HeLa cells

    • Following azetidine treatment, additional bands in the 50-70 kDa range appear

    • Tumor cell lines may show constitutive expression of the 50 kDa cleaved form

  • Troubleshooting approaches:

    • Use fresh samples with complete protease inhibitor cocktails

    • Optimize sample loading (typically 20-50 μg total protein)

    • Ensure complete denaturation and reduction (boil samples in loading buffer containing DTT or β-mercaptoethanol)

    • Test different blocking agents (milk vs. BSA)

    • Include appropriate positive controls (e.g., ATF6 transfected cell lysate)

    • Consider maximum sensitivity ECL substrate for clearer detection

Understanding the expected banding patterns and their biological significance is crucial for accurate interpretation of experimental results, particularly when studying stress response dynamics.

What are common pitfalls in ATF6 immunohistochemistry and how can they be addressed?

Immunohistochemical detection of ATF6 in tissue samples presents several technical challenges that researchers should anticipate and address:

  • Epitope accessibility issues:

    • Problem: Formalin fixation can mask ATF6 epitopes

    • Solution: Optimize antigen retrieval methods using either TE buffer pH 9.0 (recommended) or citrate buffer pH 6.0 as alternatives

    • Approach: Perform side-by-side comparisons of different retrieval methods on serial sections

  • Background staining challenges:

    • Problem: Non-specific binding causing high background

    • Solution: Optimize blocking conditions (duration, blocking agent concentration)

    • Approach: Test different blocking agents (normal serum, BSA, commercial blocking reagents)

  • Signal intensity variation:

    • Problem: Weak or inconsistent staining

    • Solution: Titrate antibody concentrations (recommended range 1:250-1:1000)

    • Approach: Include positive control tissues (human prostate tissue has been validated)

  • Specificity concerns:

    • Problem: Distinguishing specific from non-specific staining

    • Solution: Include appropriate negative controls (isotype control antibodies)

    • Approach: Consider dual staining with organelle markers to confirm subcellular localization

  • Cell type identification:

    • Problem: Difficulty identifying ATF6-positive cell types in heterogeneous tissues

    • Solution: Perform dual IHC with cell type-specific markers

    • Approach: Use serial sections or multiplex IHC approaches

  • Technical protocol considerations:

    • Recommended detection system: Anti-Rabbit IgG VisUCyte™ HRP Polymer Antibody has been validated

    • Chromogen selection: DAB (brown) with hematoxylin counterstain (blue) works well for distinguishing cytoplasmic and nuclear staining

    • Tissue preparation: Heat-induced epitope retrieval using appropriate retrieval reagents is critical

  • Interpretation guidelines:

    • Normal pattern: In human prostate, specific staining localizes to cytoplasm and nuclei

    • Stress conditions: Expect increased nuclear localization

    • Quantification: Consider digital image analysis for objective quantification

By addressing these common pitfalls proactively, researchers can generate more reliable and reproducible IHC data for ATF6 in tissue samples.

How can researchers troubleshoot inconsistent ATF6 activation patterns across different cell lines?

Inconsistent ATF6 activation patterns across cell lines reflect biological variability and technical challenges that require systematic troubleshooting:

  • Biological factors contributing to variability:

    • Basal UPR activation: Some cell lines (particularly cancer lines) may have constitutively activated UPR pathways

    • ATF6 processing efficiency: Cell-specific differences in proteolytic machinery (S1P and S2P proteases)

    • Stress sensitivity: Variable thresholds for ER stress activation between cell types

    • Isoform expression: Differential expression of ATF6α versus ATF6β

    • Genetic alterations: Mutations or polymorphisms affecting ATF6 processing or antibody epitopes

  • Technical factors affecting consistency:

    • Culture conditions: Confluence, passage number, and serum batches affect stress responses

    • Stress induction protocols: Timing, dosage, and choice of stressors impact activation patterns

    • Sample preparation: Timing between stress induction and sample collection is critical

    • Detection methods: Western blot versus immunofluorescence may yield different insights

  • Documented cell line variations:

    • Tumor cell lines like B cell lymphoma (DEL), primary effusion lymphoma (BC-3, PEL-SY, HBL-6), lymphoblastic leukemia (DS-1), and multiple myeloma (RPMI-8226, NCI-H929) show strong expression of the cleaved 50 kDa ATF6 form

    • Various cell lines validated for ATF6 antibody applications include U2OS, HeLa, HEK-293, 4T1, HSC-T6, NIH/3T3, RAW 264.7, MCF-7, Jurkat, and K-562 cells

  • Systematic troubleshooting approach:

    • Standardized culture conditions: Maintain consistent confluence, passage number, and media lots

    • Positive controls: Include cell lines with well-characterized ATF6 activation (e.g., HeLa)

    • Time course studies: Capture activation dynamics rather than single time points

    • Multiple stress inducers: Compare responses to different ER stressors (tunicamycin, thapsigargin, DTT)

    • Complementary approaches: Combine protein detection with transcriptional readouts of ATF6 targets

  • Analytical considerations:

    • Quantitative assessment: Quantify the ratio of cleaved to full-length ATF6

    • Normalization strategies: Use appropriate loading controls

    • Statistical analysis: Apply appropriate statistical methods to determine significance of observed differences

    • Validation with orthogonal methods: Confirm protein-level changes with mRNA analysis of ATF6 target genes

By systematically addressing these factors, researchers can better understand whether observed differences represent true biological variation in ATF6 regulation or technical artifacts requiring further optimization.

How might single-cell approaches enhance our understanding of ATF6 activation heterogeneity?

Single-cell methodologies offer powerful new approaches to investigate heterogeneity in ATF6 activation that is masked in population-level analyses:

  • Single-cell technologies applicable to ATF6 research:

    • Single-cell RNA-seq: Can reveal transcriptional differences in ATF6 target genes

    • Mass cytometry (CyTOF): Allows simultaneous detection of multiple UPR components

    • Live-cell imaging: Enables tracking of ATF6 localization in individual cells over time

    • Single-cell Western blotting: Provides protein-level data at single-cell resolution

    • Microfluidic platforms: Allow precise control of microenvironment and real-time monitoring

  • Key research questions addressable with single-cell approaches:

    • Cell-to-cell variability in ATF6 activation thresholds

    • Temporal dynamics of activation at the single-cell level

    • Correlation between ATF6 activation and cell fate decisions

    • Identification of distinct cellular subpopulations with different ATF6 responses

    • Integration of ATF6 signaling with other UPR branches at single-cell resolution

  • Technical considerations for implementation:

    • Antibody validation at single-cell level (specificity in flow cytometry is already established)

    • Development of ATF6 activity reporters compatible with single-cell readouts

    • Integration of subcellular localization data with activation status

    • Computational approaches for analyzing high-dimensional single-cell data

  • Potential experimental designs:

    • Single-cell RNA-seq following stress induction to identify ATF6-responsive cell clusters

    • Time-lapse imaging of fluorescently-tagged ATF6 to track individual cell responses

    • Combined flow cytometry for cleaved ATF6 and apoptosis markers to link activation to fate

    • Single-cell ChIP-seq to examine ATF6 binding patterns across individual cells

These approaches would help address fundamental questions about why some cells succumb to ER stress while others adapt and survive, potentially revealing new therapeutic avenues for diseases involving dysregulated ER stress responses.

What roles might ATF6 play in disease-specific contexts beyond the canonical UPR?

Emerging research suggests ATF6 functions extend beyond canonical UPR signaling, with implications for various disease contexts:

  • Cancer-specific roles:

    • Constitutive activation: Multiple tumor cell lines show high expression of the cleaved 50 kDa ATF6 form

    • Tumor types with documented ATF6 involvement include:

      • B cell lymphoma

      • Primary effusion lymphoma

      • Lymphoblastic leukemia

      • Multiple myeloma

    • Research implications: Investigate ATF6 as a potential therapeutic target in these malignancies

  • Neurodegenerative diseases:

    • ATF6 may play protective roles in protein misfolding diseases

    • Research opportunities:

      • Examine ATF6 activation patterns in disease models using validated antibodies

      • Investigate cell type-specific responses in CNS (neurons vs. glia)

      • Explore temporal dynamics of activation during disease progression

  • Metabolic disorders:

    • ATF6 links ER stress to metabolic regulation

    • Research directions:

      • Tissue-specific activation patterns in metabolic disease models

      • Cross-talk with other metabolic signaling pathways

      • Nutritional regulation of ATF6 processing

  • Inflammatory conditions:

    • UPR activation interfaces with inflammatory signaling

    • Investigation approaches:

      • ATF6 activation in immune cell populations

      • Regulation of inflammatory gene expression by ATF6

      • Impact of inflammatory mediators on ATF6 processing

  • Methodological considerations for disease-specific research:

    • Tissue-specific optimization of antibody protocols

    • Integration of animal models with human tissue studies

    • Consideration of ATF6α vs. ATF6β functions in disease contexts

    • Development of disease-relevant cell and organoid models

Understanding these non-canonical roles requires precise detection methodologies and careful experimental design, with ATF6 antibodies serving as essential tools for mapping activation patterns across different disease contexts.

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.