NR6A1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to NR6A1 Antibody

NR6A1 antibodies are monoclonal or polyclonal reagents that bind specifically to the NR6A1 protein, an orphan nuclear receptor encoded by the NR6A1 gene (UniProt: Q15406). This receptor regulates transcriptional networks by binding DR0 DNA motifs (AGGTCA repeats), impacting processes such as:

  • Neural crest cell (NCC) specification and survival

  • Repression of pluripotency factors (Oct4, Nanog)

  • Germ cell development and gametogenesis

These antibodies are widely used in developmental biology and stem cell research to map NR6A1 expression and function.

Neural Crest Cell Development

NR6A1 antibodies were pivotal in identifying the receptor’s role in NCC specification. Key findings include:

  • Loss-of-function phenotypes: Nr6a1 knockout mice exhibit cranial NCC deficiencies and agenesis of caudal migratory NCC due to disrupted EMT (Snail1, Zeb2) and NCC specifiers (Sox9, Sox10) .

  • Mechanistic insights: ChIP assays using NR6A1 antibodies confirmed direct binding to DR0 motifs in Sox9, Sox10, Snai1, and Zeb2 promoters, linking NR6A1 to transcriptional activation of NCC genes .

Pluripotency Regulation

  • NR6A1 represses Oct4 and Nanog by binding their promoters, as shown via competitive EMSA and ATAC-seq .

  • Overexpression of Oct4 in gastrulating embryos disrupts NCC formation, highlighting NR6A1’s role in transitioning cells from pluripotency to differentiation .

Germ Cell Development

  • NR6A1 antibodies detect expression in testicular germ cells, supporting its role in regulating BMP-15 and Gdf-9 during gametogenesis .

Western Blotting

  • Sample preparation: Lysates from HEK293, K562, or NCC-differentiated stem cells .

  • Results: Distinct bands at ~54 kDa (predicted MW) and cross-reactivity with transfected cells (Figure 1–3 in ).

Immunofluorescence

  • Protocol: Fixation with paraformaldehyde, permeabilization with Triton X-100, and staining with NR6A1 antibody (1:400 dilution) .

  • Outcome: Nuclear localization in HepG2 cells (Figure 4 in ).

Chromatin Immunoprecipitation

  • Validated in mouse ESCs to confirm NR6A1 binding to Oct4, Nanog, and NCC-specifier gene promoters .

Functional Insights from Antibody-Based Studies

Target PathwayRegulatory Role of NR6A1Experimental Evidence
NCC SpecificationActivates Sox9, Sox10, Snai1ChIP, ATAC-seq, EMSA
Pluripotency ExitRepresses Oct4, NanogKnockout models, overexpression
Axial ElongationModulates Hox gene expressionTransgenic mouse models

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Stored at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receiving them. Delivery times may vary depending on the purchasing method or location. Please contact your local distributor for specific delivery times.
Synonyms
GCNF 1 antibody; GCNF antibody; GCNF1 antibody; Germ cell nuclear factor antibody; hGCNF antibody; hRTR antibody; NR 61 antibody; NR61 antibody; nr6a1 antibody; NR6A1_HUMAN antibody; Nuclear receptor subfamily 6 group A member 1 antibody; Orphan nuclear receptor NR6A1 antibody; Retinoic acid receptor related testis associated receptor antibody; Retinoid receptor related testis specific receptor antibody; Retinoid receptor-related testis-specific receptor antibody; RTR antibody
Target Names
Uniprot No.

Target Background

Function
NR6A1 is an orphan nuclear receptor that binds to a response element containing the sequence 5'-TCAAGGTCA-3'. It is believed to play a role in regulating gene expression during germ cell development in gametogenesis.
Gene References Into Functions
  1. GCNF acts as a transcriptional repressor in the regulation of OCT4 gene expression through cooperative interaction with three NR binding elements in pluripotent NCCIT cells. PMID: 29057499
  2. GCNF demonstrated a regulatory gene pattern that is distinct from RA treatment during hES cell differentiation. It was down-regulated by 36% in RA-treated cells compared to undifferentiated hES cells, and up-regulated 64% in undifferentiated hES cells. PMID: 26769970
  3. Findings suggest a role for Gcnf as a regulator of iPS cell pluripotency gene expression. It also demonstrates that reactivation of the Gcnf gene may serve as a marker to distinguish fully reprogrammed iPS cells from incompletely pluripotent cells. PMID: 23495137
  4. Our results indicate that cellular levels of NR6A1 are correlated with disease progression in prostate cancer. PMID: 23532770
  5. These experiments confirmed that NR6A1 functions as a regulator for hypocretin transcription. PMID: 21056546
  6. RTR mRNA was expressed in the human placental choriocarcinoma cell lines BeWo, JAR, and JEG-3. PMID: 11969338
  7. characterization of a novel nuclear protein, referred to as RAP80, that interacts with the retinoid-related testis-associated receptor PMID: 12080054
  8. down-regulation of CREMtau-mediated gene expression by GCNF PMID: 15456763

Show More

Hide All

Database Links

HGNC: 7985

OMIM: 602778

KEGG: hsa:2649

STRING: 9606.ENSP00000420267

UniGene: Hs.20131

Protein Families
Nuclear hormone receptor family, NR6 subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Shows highest expression in the germ cells of the adult testis.

Q&A

What is NR6A1 and why are antibodies against it important for research?

NR6A1 (Nuclear Receptor Subfamily 6 Group A Member 1) is a nuclear receptor protein involved in transcription regulation. In humans, the canonical protein has 480 amino acid residues with a molecular mass of 54.4 kDa and is localized in the nucleus. NR6A1 is also known by several synonyms including GCNF, GCNF1, RTR, hGCNF, and retinoic acid receptor-related testis-associated receptor .

Antibodies against NR6A1 are crucial for multiple research applications because:

  • They enable detection and quantification of NR6A1 in various experimental systems

  • They allow visualization of subcellular localization patterns

  • They facilitate investigations into NR6A1's role in transcriptional regulation

  • They support studies of NR6A1's interactions with target DNA sequences

  • They help elucidate NR6A1's involvement in developmental processes and disease mechanisms

What are the principal applications of NR6A1 antibodies in molecular biology research?

NR6A1 antibodies support multiple experimental applications with specific methodological considerations:

ApplicationMethodological RecommendationsCommon Antibody Types
Western Blot (WB)Most commonly used application; recommended dilutions vary by supplier (typically 1:500-1:2000)Both monoclonal and polyclonal
Immunohistochemistry (IHC)Effective for both paraffin-embedded sections (IHC-P) and frozen sectionsPolyclonal preferred for tissue detection
Immunofluorescence (IF)Used to visualize nuclear localization of NR6A1 in fixed cellsBoth types; monoclonal for specific epitopes
ChIP-PCRCritical for studying NR6A1 binding to promoter regionsHigh-affinity monoclonal antibodies
ELISAQuantitative detection of NR6A1 protein levelsEither type depending on specificity needs
Flow Cytometry (FACS)Analysis of NR6A1 expression in cell populationsMonoclonal preferred for standardization

For Western blot applications, different antibodies target specific regions of NR6A1, with many recognizing the ligand binding domain or specific amino acid sequences (e.g., AA 65-118) .

How should researchers optimize ChIP-PCR protocols when using NR6A1 antibodies?

Chromatin Immunoprecipitation (ChIP) with NR6A1 antibodies requires specific methodological considerations:

  • Cross-linking optimization:

    • Use 1% formaldehyde for cross-linking (10-15 minutes at room temperature)

    • Quench with 0.125M glycine

    • Prepare nuclear extract according to established protocols

  • DNA fragmentation:

    • Digest genomic DNA using Micrococcal Nuclease to obtain fragments ranging from 150-900 bp

    • Confirm fragment sizes via gel electrophoresis

  • Immunoprecipitation:

    • Use 10μg of anti-GCNF monoclonal antibody for optimal capture of NR6A1 protein

    • Incubate with Protein-G agarose beads

    • Wash NR6A1-antibody-Protein-G complexes thoroughly

  • PCR amplification:

    • Design primers targeting putative NR6A1-binding sites (particularly DR0 elements and NurRE motifs)

    • Include controls with normal rabbit serum and water without DNA template

  • Validation strategy:

    • Quantitative RT-PCR analysis in cells transfected with NR6A1 expression vectors to confirm increased binding to target regions

Note that the binding capacity of NR6A1 to specific DNA motifs like NurRE may be relatively low, requiring optimization of antibody concentration and incubation conditions .

What approaches are recommended for visualizing NR6A1 expression in tissue sections?

For effective immunohistochemical detection of NR6A1 in tissue sections, researchers should follow these methodological steps:

  • Tissue preparation:

    • For paraffin sections: Fix tissue in 4% paraformaldehyde in 0.1M phosphate buffer (pH 7.4)

    • Post-fix appropriately before cryosectioning into 30-μm sections

    • For formalin-fixed paraffin-embedded tissues, prepare 4-μm sections on pre-cleaned charged microscope slides

  • Antigen retrieval:

    • Heat slides in a tissue-drying oven (45 minutes at 60°C)

    • Deparaffinize with three 5-minute washes in xylene at room temperature

  • Double immunohistochemical staining protocol:

    • Incubate sections with rabbit anti-HCRT-1 antiserum (1:5000) and mouse anti-GCNF monoclonal antibody (1:500)

    • Use secondary antibodies such as goat Alexa594-labeled anti-mouse IgG and goat Alexa488-labeled anti-rabbit IgG (1:2000)

    • Visualize under a fluorescence microscope equipped with a digital camera

  • Specificity controls:

    • Include negative controls (e.g., MOLT-4 human acute lymphoblastic leukemia cell line)

    • Include positive controls (e.g., HepG2 human hepatocellular carcinoma cell line)

    • Counter-stain nuclei with DAPI to confirm nuclear localization

The recommended antibody concentration for immunohistochemistry is typically 4-5 μg/mL .

How can researchers evaluate DNA binding properties of NR6A1 using antibodies?

To study NR6A1's interaction with DNA, researchers can implement these methodological approaches:

  • Electrophoretic Mobility Shift Assay (EMSA):

    • Prepare radiolabeled oligonucleotide probes containing direct repeat with zero spacing (DR0) of the consensus sequence AGGTCA

    • Incubate with native or mutated NR6A1 proteins

    • Verify binding specificity through competition assays using unlabeled DR0 sequences

    • Confirm NR6A1 presence in the complex via supershift assays with anti-tag antibodies (e.g., anti-Myc)

  • Competitive inhibition assays:

    • Introduce wildtype and mutated DR0 site DNA in increasing amounts

    • Observe diminished binding with increasing wildtype DNA compared to mutated sequences

    • This confirms selective binding to specific DNA motifs

  • Targeted ChIP assays:

    • Differentiate mouse embryonic stem cells (mESC) into neural crest cells (NCC)

    • Evaluate binding to DR0 sites in genes of interest (e.g., Sox9, Sox10, Snai1, Zeb2)

    • Also assess binding to other known targets like Nanog DR0 sites

These approaches can reveal how variants in NR6A1 affect DNA binding affinity, with implications for understanding developmental disorders linked to NR6A1 mutations .

What methodologies are effective for studying NR6A1's role in transcriptional regulation?

To investigate NR6A1's function in regulating gene expression, researchers should consider:

  • Chromatin accessibility analysis:

    • Compare chromatin accessibility of target genes (e.g., Oct4) in NR6A1-/- mutant embryos versus controls

    • Identify peaks showing increased or decreased accessibility of open chromatin

    • Align these with locations of putative DR0 binding sites

  • Reporter gene assays:

    • Construct reporter plasmids containing promoter regions with NR6A1 binding sites

    • Co-transfect with NR6A1 expression vectors (wildtype or mutant)

    • Measure transcriptional activity changes to determine repressive or activating functions

  • Gene expression analysis after NR6A1 manipulation:

    • Perform temporal deletion studies using conditional knockout models

    • Analyze expression of target genes using RT-qPCR

    • Evaluate changes in regulatory networks (e.g., pluripotency factors, neural crest specifiers)

  • In vivo overexpression studies:

    • Design temporal initiation of Oct4 overexpression to correlate with the temporal requirement for Nr6a1

    • Assess disruption of neural crest cell specification and formation

These approaches have revealed NR6A1's dual role as a repressor of pluripotency factors and an activator of gene regulatory networks involved in neural crest cell specification .

What are common challenges with NR6A1 antibodies and how can they be addressed?

ChallengePossible CausesRecommended Solutions
Low signal in Western blotInsufficient protein, degradation, or epitope masking1. Increase antibody concentration
2. Optimize sample preparation
3. Try different antibodies targeting different epitopes
4. Use fresh lysates and add protease inhibitors
Non-specific bindingCross-reactivity with similar proteins1. Increase blocking time/concentration
2. Optimize antibody dilution
3. Select antibodies with validated specificity
4. Use monoclonal antibodies for higher specificity
Poor immunohistochemistry stainingInadequate antigen retrieval or fixation issues1. Test different antigen retrieval methods
2. Optimize fixation time
3. Try different antibody clones
4. Use amplification systems for weak signals
Inconsistent ChIP resultsVariability in crosslinking or chromatin shearing1. Carefully control crosslinking times
2. Optimize sonication parameters
3. Confirm antibody lot-to-lot consistency
4. Include appropriate controls

When troubleshooting, remember that the binding capacity of NR6A1 to certain DNA motifs like NurRE may be naturally low , which might necessitate optimizing experimental conditions or using more sensitive detection methods.

How should researchers select the most appropriate NR6A1 antibody for specific applications?

When selecting NR6A1 antibodies, consider these methodological factors:

  • Target epitope considerations:

    • Ligand Binding Domain antibodies: Suitable for studying NR6A1's function in transcriptional regulation

    • N-terminal region antibodies (AA 30-59): Useful for detecting full-length protein

    • Mid-region antibodies: Can detect most isoforms and fragments

  • Application-specific selection:

    • For Western blot: Both monoclonal and polyclonal antibodies work well; consider epitope accessibility after denaturation

    • For ChIP: High-affinity antibodies recognizing native conformation are essential

    • For immunohistochemistry: Polyclonal antibodies often provide stronger signals in tissues

  • Species cross-reactivity needs:

    • Wide cross-reactivity (human, mouse, rat, etc.): Some antibodies show reactivity across species (up to 100% sequence identity)

    • Species-specific detection: Choose antibodies validated for specific species

  • Validation status:

    • Prioritize antibodies with published citations

    • Consider antibodies validated by multiple techniques (WB, IHC, IF)

    • Check if specificity has been confirmed by knockout/knockdown controls

For ChIP applications specifically, monoclonal antibodies like anti-GCNF (Perseus Proteomics, PP-H7921-00) have been successfully used in published research .

How are NR6A1 antibodies being used to investigate developmental biology and disease mechanisms?

Recent research has employed NR6A1 antibodies in several cutting-edge applications:

  • Neural crest cell (NCC) development studies:

    • NR6A1 antibodies help visualize expression patterns during embryonic development

    • They reveal NR6A1's role in repressing pluripotency factors while activating NCC specification genes

    • Immunohistochemistry with NR6A1 antibodies shows temporal-spatial expression patterns critical for understanding developmental timing

  • Investigation of congenital disorders:

    • NR6A1 variants have been linked to renal, uterine, and costovertebral defects

    • Antibodies enable detection of mutant NR6A1 proteins with decreased DNA binding affinity

    • They help characterize how heterozygous loss-of-function variants affect development

  • TGF-β signaling pathway research:

    • Studies of lnc-Nr6a1 as a reservoir of miR during epithelial-mesenchymal transition (EMT)

    • Expression levels of lnc-Nr6a1 isoforms and SIP1 measured by RT-qPCR analysis

    • Antibodies help track the regulation and function of NR6A1 in these processes

  • Transcriptional regulation of hypocretin/orexin:

    • NR6A1 antibodies in ChIP-PCR studies demonstrated binding to the prepro-hypocretin promoter

    • Double immunohistochemical staining revealed co-expression patterns

    • These studies have implications for understanding sleep regulation mechanisms

What advanced techniques combine NR6A1 antibodies with other methodologies for deeper mechanistic insights?

Researchers are integrating NR6A1 antibodies into sophisticated multi-modal approaches:

  • Multiomics integration:

    • Combining ChIP-seq with ATAC-seq to correlate NR6A1 binding with chromatin accessibility changes

    • Integrating RNA-seq data to connect binding events with transcriptional outcomes

    • These approaches reveal how NR6A1 modulates the chromatin landscape during development

  • iDRIP-MS (Identification of Direct RNA-Interacting Proteins by Mass Spectrometry):

    • Reveals direct lnc-Nr6a1-1-interacting proteins

    • Complements antibody-based studies by identifying protein interaction networks

    • Efficiency of lnc-Nr6a1-1 pulldown verified by RT-qPCR against standard curves

  • Combined in vivo and in vitro approaches:

    • Using human induced pluripotent stem cells (iPSCs) differentiated into NCCs alongside mouse models

    • Antibodies facilitate tracking of NR6A1 expression across different experimental systems

    • This enables translation between model systems and human development

  • Conditional spatiotemporal deletion models:

    • Global temporal and conditional spatiotemporal deletion reveals specific requirements for NR6A1

    • Antibodies confirm deletion efficiency and help track developmental consequences

    • These approaches have revealed that NCC specification in mouse embryos commences earlier than previously recognized

By employing these integrated approaches, researchers are uncovering NR6A1's complex roles in development and disease, establishing it as a novel regulator of mammalian NCC specification and formation .

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.