NR1H4 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
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchase method or location. Please contact your local distributors for specific delivery times.
Synonyms
BAR antibody; Bile acid receptor antibody; Farnesoid X activated receptor antibody; Farnesoid X receptor antibody; Farnesoid X-activated receptor antibody; Farnesol receptor HRR 1 antibody; Farnesol receptor HRR-1 antibody; Farnesol receptor HRR1 antibody; FXR antibody; HRR 1 antibody; HRR1 antibody; NR1H4 antibody; NR1H4_HUMAN antibody; Nuclear receptor subfamily 1 group H member 4 antibody; Retinoid X receptor interacting protein 14 antibody; Retinoid X receptor-interacting protein 14 antibody; RIP 14 antibody; RIP14 antibody; RXR interacting protein 14 antibody; RXR-interacting protein 14 antibody
Target Names
NR1H4
Uniprot No.

Target Background

Function
The Farnesoid X Receptor (FXR), encoded by the NR1H4 gene, is a ligand-activated transcription factor that serves as a receptor for bile acids (BAs), including chenodeoxycholic acid (CDCA), lithocholic acid, deoxycholic acid (DCA), and allocholic acid (ACA). FXR plays a crucial role in bile acid homeostasis by regulating genes involved in bile acid synthesis, conjugation, and enterohepatic circulation. Additionally, it modulates lipid and glucose homeostasis and participates in innate immune responses.
The FXR-RXR heterodimer primarily binds to farnesoid X receptor response elements (FXREs) containing two inverted repeats of the consensus sequence 5'-AGGTCA-3' spaced by 1 nucleotide (IR-1). However, it also binds to tandem repeat DR1 sites with lower affinity and can be activated by either FXR or RXR-specific ligands.
Monomeric nuclear receptors, such as NR5A2/LRH-1, bound to coregulatory nuclear responsive element (NRE) halfsites located in close proximity to FXREs, are suggested to modulate transcriptional activity.
In the liver, FXR activates transcription of the corepressor NR0B2, indirectly inhibiting CYP7A1 and CYP8B1 (involved in bile acid synthesis). This inhibition involves, at least in part, histone demethylase KDM1A, leading to epigenomic repression. FXR also activates the transcription of SLC10A1/NTCP (involved in hepatic uptake of conjugated BAs) and the repressor MAFG (involved in regulation of bile acid synthesis). Furthermore, FXR activates the transcription of SLC27A5/BACS and BAAT (involved in bile acid conjugation), ABCB11/BSEP (involved in bile salt export) by directly recruiting histone methyltransferase CARM1, and ABCC2/MRP2 (involved in secretion of conjugated BAs) and ABCB4 (involved in secretion of phosphatidylcholine in the small intestine).
In the intestine, FXR activates FGF19 expression and secretion, leading to hepatic CYP7A1 repression. This function also involves the coordinated induction of hepatic KLB/beta-klotho expression. FXR regulates the transcription of liver UGT2B4 and SULT2A1, which are involved in bile acid detoxification. Binding to the UGT2B4 promoter suggests a monomeric transactivation independent of RXRA.
FXR modulates lipid homeostasis by activating liver NR0B2/SHP-mediated repression of SREBF1 (involved in de novo lipogenesis), expression of PLTP (involved in HDL formation), SCARB1 (involved in HDL hepatic uptake), APOE, APOC1, APOC4, PPARA (involved in beta-oxidation of fatty acids), VLDLR and SDC1 (involved in the hepatic uptake of LDL and IDL remnants), and inhibiting the expression of MTTP (involved in VLDL assembly). FXR increases the expression of APOC2, promoting lipoprotein lipase activity, which is implicated in triglyceride clearance. FXR transrepresses APOA1, involving a monomeric competition with NR2A1 for binding to a DR1 element. FXR also reduces triglyceride clearance by inhibiting the expression of ANGPTL3 and APOC3, both involved in the inhibition of lipoprotein lipase.
FXR participates in glucose homeostasis by modulating hepatic gluconeogenesis through activation of NR0B2/SHP-mediated repression of respective genes. It modulates glycogen synthesis by inducing phosphorylation of glycogen synthase kinase-3. FXR also modulates glucose-stimulated insulin secretion and is involved in insulin resistance.
FXR is involved in intestinal innate immunity, playing a role in protecting the distal small intestine against bacterial overgrowth and preserving the epithelial barrier. It down-regulates inflammatory cytokine expression in several types of immune cells, including macrophages and mononuclear cells. FXR mediates trans-repression of TLR4-induced cytokine expression. This function appears to require its sumoylation and prevents N-CoR nuclear receptor corepressor clearance from target genes, such as IL1B and NOS2. FXR is involved in the TLR9-mediated protective mechanism in intestinal inflammation. FXR plays an anti-inflammatory role in liver inflammation by inhibiting proinflammatory (but not antiapoptotic) NF-kappa-B signaling.
FXR promotes transcriptional activation of target genes NR0B2/SHP (inducible by unconjugated CDCA), SLC51B/OSTB (inducible by unconjugated CDCA and DCA) and FABP6/IBAP. It shows low activity for ABCB11/BSEP (inducible by unconjugated CDCA, DCA and ACA) and is not inducible by taurine- and glycine-amidated CDCA.
FXR promotes transcriptional activation of target genes ABCB11/BSEP (inducible by unconjugated CDCA, DCA and ACA), NR0B2/SHP (inducible by unconjugated CDCA DCA and ACA), SLC51B/OSTB (inducible by unconjugated CDCA and DCA) and FABP6/IBAP. It is not inducible by taurine- and glycine-amidated CDCA.
FXR promotes transcriptional activation of target genes NR0B2/SHP (inducible by unconjugated CDCA), SLC51B/OSTB (inducible by unconjugated CDCA and DCA) and IBAP. It shows low activity for ABCB11/BSEP (inducible by unconjugated CDCA, DCA and ACA) and is not inducible by taurine- and glycine-amidated CDCA.
FXR promotes transcriptional activation of target genes ABCB11/BSEP (inducible by unconjugated CDCA, ACA and DCA), NR0B2/SHP (inducible by unconjugated CDCA, ACA and DCA), SLC51B/OSTB (inducible by unconjugated CDCA and DCA) and FABP6/IBAP. It is the most efficient isoform compared to isoforms 1 to 3 and is not inducible by taurine- and glycine-amidated CDCA.
Gene References Into Functions
  1. FXR pathway activation increases XBP1 splicing and enhances p-IRE1alpha expression in liver PMID: 29377207
  2. Activation of FXR inhibits, whereas TGR5 activation may promote, cholangiocarcinoma progression by regulating proliferation, migration and mitochondrial energy metabolism. PMID: 28916388
  3. A study demonstrated for the first time a novel target for farnesoid X-activated receptor (FXR) and that the activated receptor alters the acquisition of sperm fertilizing ability. PMID: 29921626
  4. Low FXR expression is associated with colorectal cancer. PMID: 30106441
  5. Studies indicate that the deregulation of farnesoid X receptor (FXR) may lead to abnormalities of specific organs and metabolic dysfunction [Review]. PMID: 30013008
  6. FXR agonist treatment enhanced TGF-beta-induced epithelial mesenchymal transition(EMT) morphologic changes and FXR antagonist inhibited the effect of TGF-beta. Thus, FXR activation enhances EMT in hepatocellular carcinoma (HCC) and FXR antagonists may be EMT-suppressing drug candidates. PMID: 29958417
  7. This study provides some data suggesting the possible involvement of FXR in the pathophysiology and development of thyroid neoplasms. In particular, we found that there are differences regarding FXR expression between papillary carcinomas and hyperplastic nodules, being also correlated with some patients' clinicopathological parameters. PMID: 28762281
  8. Data suggest that TGR5 and FXR in intestinal mucosa are important for glucose homeostasis, in particular in metabolic disorders such as type 2 diabetes and obesity. (TGR5 = membrane-type receptor for bile acids TGR5; FXR = farnesoid X receptor) [REVIEW; Congress as Topic] PMID: 27846919
  9. These results suggested that FXR may serve as an important negative regulator for manipulating Smad3 expression, and the FXR/Smad3 pathway may be a novel target for the treatment of renal fibrosis. PMID: 27853248
  10. Loss of FXR or its down-regulation is associated with higher bile acids concentrations and with a pro-tumorigenic phenotype. (Review) PMID: 28400119
  11. Lys-325 is a non-canonical site of SUMOylation of human FXR. CK2 is the priming effector that phosphorylates Ser-327, resulting in enhanced SUMO2 conjugation, which then directs the ubiquitination and degradation of FXR through the recruitment of the SUMO-dependent ubiquitin E3 ligase RNF4. PMID: 28201649
  12. We proposed a model to link FXR to Sp1, which included triggered FXR, p38/MAPK and/or PI3K/AKT signaling and phosphorylated Sp1, to illustrate the potential crosstalk between the two factors. PMID: 28402278
  13. The presented evidence suggested that WA can inhibit HCC cell proliferation and tumorigenesis through miR-22-repressed CCNA2, which was at least partially through FXR regulation. PMID: 27738335
  14. The results indicated that epigenetically regulated miR-449a targets CREB5 to increase FXRalpha expression, thereby promoting HBV replication and gene expression. Our findings provide a new understanding of the role of miRNAs in HBV replication. PMID: 27138288
  15. In diabetic humans, there is decreased FXR expression in the kidney. FXR plays an important role in Diabetic Kidney Disease. PMID: 27045028
  16. FXR regulates serum triglyceride level in part through PLA2G12B. PMID: 27471003
  17. It regulates the growth of renal adenocarcinoma cells. PMID: 28496032
  18. We conclude that the FXR-Gank-TSPs-Stem cells pathway is a key determinant of liver cancer in animal models and in pediatric liver cancer. Our data provide a strong basis for the development of FXR-Gank-based therapy for the treatment of patients with hepatoblastoma. PMID: 28535186
  19. PPARalpha and FXR function coordinately to integrate liver energy balance. PMID: 28287408
  20. These studies investigated the effects of FXR activation on HBV replication and of infection on the FXR pathway in differentiated HepaRG cells and in primary human hepatocytes (PHHs). PMID: 27251172
  21. Treatment with LXR and FXR ligands initiates coated platelet formation, which is thought to support coagulation but results in desensitization to platelet stimuli through inhibition of alphaIIbbeta3 consistent with their ability to inhibit platelet function and stable thrombus formation in vivo. PMID: 28619996
  22. FXR ligands inhibit platelet activation. PMID: 27758768
  23. Taken together, our data provide the first evidence that FXR suppresses proliferation of human liver cancer cells via the inhibition of the mTOR/S6K signaling pathway. FXR expression can be used as a biomarker of personalized mTOR inhibitor treatment assessment for liver cancer patients. PMID: 27109477
  24. Activated FXR inhibits leptin signaling and counteracts tumor-promoting activities of cancer-associated fibroblasts in breast malignancy. PMID: 26899873
  25. FXR may promote the proliferation of tumor cells and the hepatocytes in the process of liver regeneration by activating the PDK4-mediated metabolic reprogramming to generate glycolytic intermediates essential for rapid biomass generation, establishing a mechanistic link between cell proliferation and metabolic switch. PMID: 26728993
  26. This is the first report of bile acid derivatives able to antagonize GPBAR1 and farnesoid X receptor (FXR) modulatory activity. PMID: 26607331
  27. Induction of SOCS3 may be a novel mechanism by which FXR exerts its anti-hepatocellular carcinoma effects. PMID: 26416445
  28. Mutations in NR1H4 cause progressive familial intrahepatic cholestasis. PMID: 26888176
  29. We drew a conclusion that curcumin attenuated Alcoholic liver disease by modulating lipid deposition in hepatocytes via a Nrf2/FXR activation-dependent mechanism. PMID: 26305715
  30. miR-122 is a novel target gene of FXR, and the upregulation of miR-122 by FXR represses the growth of hepatocellular carcinoma cells, suggesting that FXR may serve as a key transcriptional regulator for manipulating miR-122 expression. PMID: 26302777
  31. IL-18 repressed FXR mRNA and protein levels in HepG2 cells. PMID: 26292095
  32. FXR signaling in ileum biopsies of humans positively correlates with body mass index. PMID: 26670557
  33. FXR activation in L cells decreases proglucagon expression by interfering with the glucose-responsive factor Carbohydrate-Responsive Element Binding Protein (ChREBP) and GLP-1 secretion by inhibiting glycolysis. PMID: 26134028
  34. FXR is decreased in obese animal models associated with INS resistance. Regulation of bile acids and FXR in the stage of obesity may prevent more metabolism disorders. PMID: 26488943
  35. These results provide important information to prioritize chemicals for further investigation and suggest possible modes of action of compounds in FXR signaling. PMID: 25257666
  36. FXR may regulate SOD3 expression to suppress reactive oxygen species production, resulting in decreasing JNK activity. PMID: 25496033
  37. Liver nuclear receptors, FXR and SHP, and bile acid transporters, NTCP and BSEP, are associated with the progression of NAFLD. PMID: 26019035
  38. Three FXR gene variants (rs35724, rs11110385, rs11110386) were identified as potential susceptibility factors for cholelithiasis in a German cohort in gender- and weight-dependent manners. PMID: 25242139
  39. These data demonstrate that copper-mediated nuclear receptor dysfunction disrupts liver function in WD and potentially in other disorders associated with increased hepatic copper levels. PMID: 26241054
  40. FXR agonist, GW4064, upregulates adipokine expression in preadipocytes and HepG2 cells. PMID: 25400456
  41. NHR1H4 genotype increases the risk for spontaneous bacterial peritonitis in live cirrhosis patients. PMID: 25086996
  42. Results show that HRG is a novel transcriptional target gene of FXR in human hepatoma cells, human upcyteVR primary hepatocytes and 3D human liver microtissues in vitro and in mouse liver in vivo. PMID: 25363753
  43. Established genome-wide human FXR binding and transcriptome profiles. These results will aid in determining the human FXR functions, as well as judging to what level the mouse models could be used to study human FXR functions. PMID: 25198545
  44. Our data showed that bile acid-activated FXR stimulates miR-22-silenced CCNA2, a novel pathway for FXR to exert its protective effect in the gastrointestinal tract. PMID: 25596928
  45. AB23A produces a protective effect against ANIT-induced hepatotoxicity and cholestasis, due to FXR-mediated regulation of transporters and enzymes. PMID: 25655198
  46. Compared with normal gallbladder tissues, FXR expression was decreased and MCL1 expression was increased in GBC, during progression of tumor node metastasis (TNM) stage. PMID: 25043081
  47. Intestinal nuclear bile acid farnesoid X receptor is regulated transcriptionally by CDX2. PMID: 25138215
  48. Significant differences at the distal (-1890) and proximal promoter (-358) CpG sites of the FXR/NR1H4 and at the distal PXR/NR1I2 (-1224) promoter, which were consistently less methylated in ICP cases when compared with controls. PMID: 24498169
  49. Show that FXR interacts with and is O-GlcNAcylated by O-GlcNAc transferase in its N-terminal AF1 domain. PMID: 24037988
  50. NR1H4 methylation is associated with colon cancer. PMID: 24169962

Show More

Hide All

Database Links

HGNC: 7967

OMIM: 603826

KEGG: hsa:9971

STRING: 9606.ENSP00000447149

UniGene: Hs.282735

Involvement In Disease
Cholestasis, progressive familial intrahepatic, 5 (PFIC5)
Protein Families
Nuclear hormone receptor family, NR1 subfamily
Subcellular Location
Nucleus.; [Isoform 1]: Nucleus.; [Isoform 2]: Nucleus.; [Isoform 3]: Nucleus.; [Isoform 4]: Nucleus.
Tissue Specificity
Liver and hepatocyte-related cells express mainly FXRalpha1-type isoforms with isoform 3 and isoform 4 in approximately equal proportions. In intestine and kidney mainly FXRalpha2-type isoforms are expressed with isoform 1 and isoform 2 in approximately e

Q&A

What is NR1H4 and why is it a significant research target?

NR1H4 (Nuclear Receptor Subfamily 1 Group H Member 4), also known as Farnesoid X Receptor (FXR), Bile Acid Receptor, HRR-1, RIP14, or Retinoid X receptor-interacting protein 14, is a nuclear receptor that functions as a transcription factor with a molecular mass of approximately 57 kDa . Its significance stems from its role as a global regulator of bile acid metabolism, controlling genes involved in bile acid biosynthesis (such as CYP7A1) and recycling (like IBABP) . Recent research has revealed its involvement in various cancer types, including clear cell Renal cell carcinoma (ccRCC), where it promotes cancer cell proliferation, migration, and invasion through the regulation of CCNE2 . As a nuclear receptor that binds to DNA as a heterodimer with Retinoid X Receptor (RXR), NR1H4 represents an important target for understanding both normal physiological processes and pathological conditions .

What are the key applications for NR1H4 antibodies in basic research?

NR1H4 antibodies serve multiple research applications, making them valuable tools for investigating this nuclear receptor's expression and function. The primary applications include:

  • Western Blot (WB): For detecting NR1H4 protein levels in tissue or cell lysates, with expected band size at approximately 56 kDa

  • Immunohistochemistry-Paraffin (IHC-P): For analyzing protein expression in fixed tissue sections

  • Immunocytochemistry/Immunofluorescence (ICC/IF): For visualizing subcellular localization in cultured cells

  • Flow Cytometry (Flow Cyt): For quantifying NR1H4 expression at the single-cell level

These techniques enable researchers to evaluate NR1H4 expression patterns across different tissues, subcellular compartments, and experimental conditions, providing crucial insights into its biological functions and pathological alterations.

How do I select the appropriate NR1H4 antibody for my specific research application?

Selection of an appropriate NR1H4 antibody should be based on several key considerations:

  • Target epitope: Determine whether you need an antibody targeting a specific region (e.g., N-terminal as in ab187735)

  • Antibody type: Consider whether monoclonal (for high specificity) or polyclonal (for broader epitope recognition) is more suitable

  • Validated applications: Review validation data for your intended application (WB, IHC, ICC/IF, Flow Cytometry)

  • Species reactivity: Ensure compatibility with your experimental model (human, mouse, etc.)

  • Published validation: Check if the antibody has been cited in peer-reviewed publications

For instance, mouse monoclonal antibodies targeting the N-terminal region (amino acids 1-50) have been validated for multiple applications with human samples . When working with specific cell lines like A549, antibodies such as the rabbit anti-NR1H4 (A00835-1) have demonstrated effectiveness in immunofluorescence and flow cytometry applications .

What are the optimal conditions for using NR1H4 antibodies in Western blot assays?

For optimal Western blot results with NR1H4 antibodies, the following protocol parameters have been validated:

  • Gel preparation: Use 5-20% SDS-PAGE gel

  • Electrophoresis conditions: Run at 70V (stacking gel) followed by 90V (resolving gel) for 2-3 hours

  • Sample loading: 30 μg of protein per lane under reducing conditions

  • Transfer conditions: Transfer proteins to nitrocellulose membrane at 150 mA for 50-90 minutes

  • Blocking: 5% non-fat milk in TBS for 1.5 hours at room temperature

  • Primary antibody: Incubate with anti-NR1H4 antibody at 0.5 μg/mL overnight at 4°C

  • Washing: TBS with 0.1% Tween, 3 times for 5 minutes each

  • Secondary antibody: Goat anti-rabbit IgG-HRP at 1:5000 dilution for 1.5 hours at room temperature

  • Detection: Use enhanced chemiluminescent detection kit

The expected molecular weight for NR1H4 is approximately 56 kDa. Different tissue samples (such as HCCT and HCCP tissue lysates) have been successfully used with this protocol .

How can I optimize immunofluorescence staining for NR1H4 localization studies?

For successful immunofluorescence detection of NR1H4, follow these methodological recommendations:

  • Cell preparation: Grow cells on appropriate coverslips or chamber slides

  • Fixation: Use 4% paraformaldehyde to preserve cellular structure

  • Permeabilization: Apply 0.1% Triton X-100 in PBS to allow antibody access to intracellular targets

  • Antigen retrieval: For some applications, perform enzyme antigen retrieval using appropriate reagents for 15 minutes

  • Blocking: Block with 10% goat serum to reduce non-specific binding

  • Primary antibody: Incubate with anti-NR1H4 antibody (2 μg/mL) overnight at 4°C

  • Secondary antibody: Use DyLight®488 Conjugated anti-species IgG at 1:100 dilution, incubate for 30 minutes at 37°C

  • Nuclear counterstain: Apply DAPI for nuclear visualization

  • Visualization: Use fluorescence microscope with appropriate filter sets

This protocol has been validated with A549 cells. For co-localization studies, consider using antibodies against known interaction partners like RXR or downstream targets such as CCNE2 .

What controls should be included when performing flow cytometry with NR1H4 antibodies?

When conducting flow cytometry experiments with NR1H4 antibodies, include the following controls to ensure reliable data interpretation:

  • Isotype control: Use species-matched IgG (e.g., rabbit IgG at 1 μg/10^6 cells) to assess non-specific binding

  • Unstained control: Include cells without primary or secondary antibody to establish autofluorescence baseline

  • Secondary-only control: Cells with secondary antibody but no primary antibody to detect non-specific secondary binding

  • Positive control: Include a cell line known to express NR1H4 (e.g., A549 cells)

  • Negative control: When possible, include NR1H4 knockout or knockdown cells

For intracellular staining, ensure proper fixation with 4% paraformaldehyde and permeabilization with appropriate buffer. Blocking with 10% normal goat serum is recommended. Typical antibody concentrations include primary antibody at 1 μg/10^6 cells and fluorochrome-conjugated secondary antibody at 5-10 μg/10^6 cells, with 30 minutes incubation at 20°C .

How can I establish NR1H4 knockout cell lines for functional studies?

Creating NR1H4 knockout cell lines for functional studies can be achieved through CRISPR/Cas9 technology following this methodological approach:

  • Vector design: Clone CRISPR/Cas9 systems targeting NR1H4 with GFP reporter proteins

  • Transfection: Transfect target cells (e.g., colon cancer cells) with the CRISPR/Cas9 construct

  • Selection: After 48 hours, sort GFP-positive cells using FACS

  • Single-cell cloning: Seed sorted cells onto 96-well plates at low density to obtain single-cell-derived colonies

  • Clone screening: Evaluate colonies by immunoblotting for NR1H4 expression

  • Validation: Verify knockout by genomic sequencing and functional assays

This approach has been successfully used to generate NR1H4 knockout colon cancer cell lines, resulting in phenotypes with impaired cell proliferation, reduced colony formation, and increased apoptotic cell death compared to control cells . Similar approaches can be adapted for other cell types based on research objectives.

What methodologies are effective for studying NR1H4's role in gene regulation?

To investigate NR1H4's role in gene regulation, several complementary methodologies can be employed:

  • RT^2 Profiler PCR array: Use specialized arrays like the Human Signal Transduction Pathway Finder kit to profile altered signaling pathways in NR1H4 knockout versus wild-type cells

  • Gene set enrichment analysis (GSEA): Apply this technique to identify pathways enriched in NR1H4 high versus low expression groups

  • Chromatin immunoprecipitation (ChIP): Use NR1H4 antibodies to identify direct DNA binding sites

  • Reporter gene assays: Construct reporters containing putative NR1H4 response elements to measure transcriptional activity

  • Quantitative real-time PCR (qRT-PCR): Measure expression changes in target genes like CYP7A1 and IBABP following NR1H4 modulation

  • RNA-seq: Perform transcriptome-wide analysis to identify global gene expression changes

Studies employing these approaches have revealed that NR1H4 regulates genes involved in bile acid metabolism and can also impact tumor-associated signaling pathways in cancer models .

How can I effectively study NR1H4 protein-protein interactions, particularly with RXR?

To investigate NR1H4 protein-protein interactions, especially its heterodimer formation with Retinoid X Receptor (RXR), consider these methodological approaches:

  • Co-immunoprecipitation (Co-IP): Use anti-NR1H4 antibodies to pull down protein complexes, followed by immunoblotting for RXR or other potential interaction partners

  • Proximity ligation assay (PLA): Detect protein interactions in situ with high sensitivity and specificity

  • Fluorescence resonance energy transfer (FRET): Tag NR1H4 and potential partners with appropriate fluorophores to detect interactions in living cells

  • Bimolecular fluorescence complementation (BiFC): Split fluorescent proteins fused to NR1H4 and interaction partners will reconstitute fluorescence when proteins interact

  • Mammalian two-hybrid assay: Use reporter constructs to detect transcriptional activity resulting from protein-protein interactions

Since NR1H4 binds to DNA only as a heterodimer with RXR , these techniques are crucial for understanding the functional relevance of this interaction in various physiological and pathological contexts.

How should I interpret discrepancies between NR1H4 mRNA and protein expression levels in my samples?

When facing discrepancies between NR1H4 mRNA and protein expression levels, consider these methodological approaches for interpretation:

  • Post-transcriptional regulation: Evaluate microRNA profiles that might target NR1H4 mRNA

  • Protein stability: Assess proteasomal degradation by treating samples with proteasome inhibitors

  • Translation efficiency: Perform polysome profiling to evaluate translation status

  • Isoform-specific detection: Ensure that your detection methods (primers or antibodies) recognize all relevant isoforms

  • Temporal dynamics: Consider time-course experiments to capture potential delays between transcription and translation

  • Spatial compartmentalization: Use subcellular fractionation to determine if protein localization affects detection

Research has shown that NR1H4 expression can be complex, with bioinformatic analyses revealing multiple levels of regulation including genetic alteration and DNA methylation that significantly impact patient prognosis in cancer studies . A comprehensive multi-omics approach may be necessary to fully understand these discrepancies.

What methodological considerations are important when evaluating NR1H4 expression in cancer tissues versus normal tissues?

When comparing NR1H4 expression between cancer and normal tissues, these methodological considerations are critical:

  • Tissue heterogeneity: Use laser capture microdissection to isolate specific cell populations

  • Sample matching: Whenever possible, use matched tumor and adjacent normal tissue from the same patient

  • Cancer subtypes: Stratify analysis by cancer subtypes, stages, and grades

  • Multiple detection methods: Combine RNA-seq, qRT-PCR, immunohistochemistry, and western blotting for comprehensive assessment

  • Quantification approach: For IHC, use standardized scoring systems (H-score, Allred score) and automated image analysis when possible

  • Statistical analysis: Apply appropriate statistical tests and corrections for multiple comparisons

Studies have shown that NR1H4 is highly expressed in ccRCC tissues compared to normal tissues, with diagnostic potential particularly for early-stage disease (area under ROC curve > 0.8) . Similar methodological approaches can be applied to investigate NR1H4 expression in other cancer types.

How can I determine whether NR1H4 plays a causative role in cancer progression rather than being merely correlative?

To establish a causative role for NR1H4 in cancer progression beyond correlation, implement these methodological approaches:

  • Genetic manipulation studies:

    • Generate NR1H4 knockout cell lines using CRISPR/Cas9

    • Create stable overexpression systems

    • Employ inducible expression systems for temporal control

  • Functional assays to assess cancer hallmarks:

    • Proliferation (EdU incorporation, MTT assay)

    • Migration and invasion (transwell and wound healing assays)

    • Apoptosis (flow cytometry with Annexin V staining)

    • Colony formation assays

  • Mechanistic investigations:

    • Identify downstream effectors (e.g., CCNE2)

    • Perform rescue experiments (restoring affected pathways)

    • Use pharmacological modulators of NR1H4 activity

  • In vivo studies:

    • Xenograft models with NR1H4-modulated cells

    • Patient-derived xenografts

    • Genetically engineered mouse models

Research has demonstrated that NR1H4 knockdown significantly suppresses cancer cell proliferation, migration, and invasion, while mechanistic studies have identified regulation of CCNE2 as a potential mechanism in ccRCC . Similar comprehensive approaches should be applied when investigating NR1H4's role in other cancer types.

What are the best approaches for studying the relationship between NR1H4 expression and immune cell infiltration in cancer?

To investigate associations between NR1H4 and immune cell infiltration in cancer, consider these methodological approaches:

  • Bioinformatic analysis:

    • Use tools like TISIDB (http://cis.hku.hk/TISIDB/index.php) to assess correlations between NR1H4 expression and tumor-infiltrating immune cells in public datasets

    • Apply deconvolution algorithms (CIBERSORT, xCell) to estimate immune cell populations from bulk RNA-seq data

  • Multiplexed immunohistochemistry/immunofluorescence:

    • Perform multispectral imaging to simultaneously detect NR1H4 and immune cell markers

    • Quantify spatial relationships between NR1H4-expressing cells and immune populations

  • Single-cell RNA sequencing:

    • Profile both cancer and immune cells to understand cell-specific expression patterns

    • Identify potential paracrine interactions

  • Functional validation:

    • Co-culture NR1H4-modulated cancer cells with immune cells

    • Assess changes in immune cell function (cytokine production, cytotoxicity)

    • Evaluate immune checkpoint molecule expression

Research has demonstrated that NR1H4 expression is associated with immune cell infiltration levels in ccRCC, suggesting potential implications for immunotherapy approaches . These methodologies can help determine whether NR1H4 directly influences the tumor immune microenvironment or if these associations are indirect.

What strategies can address weak or absent NR1H4 signal in Western blot experiments?

When encountering weak or absent NR1H4 signal in Western blots, implement these methodological solutions:

  • Sample preparation optimization:

    • Ensure complete cell lysis using appropriate buffers with protease inhibitors

    • Consider subcellular fractionation to enrich nuclear proteins

    • Optimize protein loading (50-100 μg may be needed for low abundance targets)

  • Transfer conditions:

    • Extend transfer time for large proteins

    • Consider wet transfer instead of semi-dry for better efficiency

    • Verify transfer efficiency with reversible staining (Ponceau S)

  • Antibody optimization:

    • Test different antibody concentrations (0.5-5 μg/mL)

    • Extend primary antibody incubation (overnight at 4°C or longer)

    • Try alternative NR1H4 antibodies targeting different epitopes

  • Signal enhancement:

    • Use more sensitive detection systems (enhanced chemiluminescence)

    • Consider amplification systems for low abundance proteins

    • Reduce washing stringency if signal is completely absent

Published protocols have successfully detected NR1H4 at approximately 56 kDa using specific conditions including 5-20% SDS-PAGE gels and primary antibody concentration of 0.5 μg/mL .

How can I minimize background and optimize signal-to-noise ratio in NR1H4 immunofluorescence experiments?

To improve signal-to-noise ratio in NR1H4 immunofluorescence experiments, apply these methodological approaches:

  • Fixation optimization:

    • Compare different fixatives (paraformaldehyde, methanol, acetone)

    • Minimize fixation time to prevent epitope masking

  • Blocking enhancement:

    • Use species-appropriate serum (10% goat serum)

    • Add 0.1-0.3% Triton X-100 to blocking buffer

    • Consider adding 1-5% BSA to reduce non-specific binding

  • Antibody conditions:

    • Titrate antibody concentration (starting at 2 μg/mL)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Increase washing steps (5-6 washes of 5 minutes each)

  • Advanced techniques:

    • Apply tyramide signal amplification for weak signals

    • Use confocal microscopy to reduce out-of-focus background

    • Consider spectral unmixing for autofluorescence removal

Validated protocols have successfully used anti-NR1H4 antibodies at 2 μg/mL with overnight incubation at 4°C and DyLight®488 conjugated secondary antibodies at 1:100 dilution for 30 minutes at 37°C .

What approaches can validate antibody specificity for NR1H4 in research applications?

To ensure antibody specificity for NR1H4, implement these methodological validation approaches:

  • Genetic validation:

    • Test antibody in NR1H4 knockout or knockdown models

    • Compare with NR1H4 overexpression systems

    • Use siRNA-mediated silencing of NR1H4 as control

  • Peptide competition:

    • Pre-incubate antibody with immunizing peptide

    • Compare staining pattern with and without peptide competition

  • Multiple antibody validation:

    • Use different antibodies targeting distinct epitopes

    • Compare detection patterns across techniques (WB, IF, IHC)

  • Cross-reactivity assessment:

    • Test specificity against closely related nuclear receptors

    • Perform immunoprecipitation followed by mass spectrometry

  • Published validation evidence:

    • Review existing literature using the same antibody

    • Check antibody citation records in research publications

Rigorous validation is especially important for nuclear receptors like NR1H4 that may share structural similarities with other family members.

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 2024 Thebiotek. All Rights Reserved.