NR1H2 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

NR1H2 (also called LXRB) is a liver X receptor beta (LXRB) that forms heterodimers with retinoid X receptors (RXRs) to regulate genes involved in cholesterol homeostasis, inflammation, and macrophage function . The NR1H2 antibody specifically detects this protein in experimental settings, enabling researchers to study its expression patterns and mechanistic roles.

Primary Uses

  • Western Blot: Detects NR1H2 in human and pig liver tissues .

  • Functional Studies: Used to investigate NR1H2's role in cholesterol uptake via MYLIP-dependent ubiquitination of LDLR and VLDLR .

  • Disease Models: Applied in atherosclerosis, prostate cancer, and nonalcoholic steatohepatitis (NASH) research .

Recommended Dilutions

ApplicationDilution Range
Western Blot1:500 – 1:2000
IHC/ChIP/ELISASample-dependent; requires titration

Table: Selected Studies Using NR1H2 Antibody

Study Title (Simplified)Key InsightCitation
EGFR/FOXO3A/LXR-α Axis in Prostate CancerNR1H2 activation promotes cancer metastasis via lipid metabolism pathways
TL1A Inhibits AtherosclerosisNR1H2 modulates vascular smooth muscle cell phenotypes
NR1H2 in Blastoid FormationNR1H2 activation enhances embryonic stem cell differentiation efficiency
Diosmetin in NASH TreatmentNR1H2-mediated pathways reduce hepatic inflammation

Validation and Quality Control

  • Specificity: Confirmed via knockdown (KD) and knockout (KO) models in hepatic studies .

  • Reproducibility: Validated in wild-type and hLXR knockin mouse liver lysates .

  • Storage: Stable at -20°C in PBS with 50% glycerol and 0.02% sodium azide .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery time estimates.
Synonyms
Liver X nuclear receptor beta antibody; Liver X receptor beta antibody; LX receptor beta antibody; LXR b antibody; LXR beta antibody; LXRB antibody; NER antibody; NER I antibody; NER1 antibody; NR1H 2 antibody; NR1H2 antibody; NR1H2_HUMAN antibody; Nuclear orphan receptor LXR beta antibody; Nuclear receptor NER antibody; Nuclear receptor subfamily 1 group H member 2 antibody; OR-1 antibody; Oxysterols receptor LXR-beta antibody; RIP15 antibody; Steroid hormone nuclear receptor NER antibody; Ubiquitously-expressed nuclear receptor antibody; UNR antibody
Target Names
NR1H2
Uniprot No.

Target Background

Function
The NR1H2 protein is a nuclear receptor exhibiting ligand-dependent transcriptional activation activity. It preferentially binds to double-stranded oligonucleotide direct repeats with the consensus half-site sequence 5'-AGGTCA-3' and a 4-nt spacing (DR-4). NR1H2 regulates cholesterol uptake through MYLIP-dependent ubiquitination of LDL receptor (LDLR), very low-density lipoprotein receptor (VLDLR), and low-density lipoprotein receptor-related protein 8 (LRP8). It also functionally interacts with RORA to regulate genes involved in liver metabolism. NR1H2 induces LPCAT3-dependent phospholipid remodeling in endoplasmic reticulum (ER) membranes of hepatocytes, driving SREBF1 processing and lipogenesis. Through LPCAT3, NR1H2 triggers the incorporation of arachidonate into phosphatidylcholines of ER membranes, increasing membrane dynamics and enabling triacylglycerol transfer to nascent very low-density lipoprotein (VLDL) particles. Additionally, via LPCAT3, NR1H2 counteracts lipid-induced ER stress response and inflammation, potentially by modulating SRC kinase membrane compartmentalization and limiting the synthesis of lipid inflammatory mediators. NR1H2 plays an anti-inflammatory role during the hepatic acute phase response by acting as a corepressor, inhibiting the hepatic acute phase response by preventing the dissociation of the N-Cor corepressor complex.
Gene References Into Functions
  1. In macrophages infected with Mycobacterium tuberculosis, IL-36 signaling modulates cholesterol biosynthesis and efflux via LXR. PMID: 29367626
  2. Research suggests that in gestational diabetes mellitus, cellular cholesterol homeostasis in the fetoplacental endothelium is modulated through LXR activation, contributing to its proper functionality. PMID: 29778664
  3. These findings provide fundamental information for evaluating the effectiveness of LXR-targeted treatment in refractory carcinoma tissues of the human lung and pancreas. PMID: 29181837
  4. LXR gene expression was significantly elevated in obese children with obstructive sleep apnea-hypopnea syndrome (OSAHS). The severity of OSAHS showed a positive correlation with COX-2 levels. PMID: 28676625
  5. ULK1 plays a role in RPS6KB1-NCOR1 repression of NR1H/LXR-mediated Scd1 transcription, exacerbating lipotoxicity in hepatic cells. PMID: 27846372
  6. These data indicate that high doses of insulin downregulate apoA-I gene expression in HepG2 cells through the redistribution of FOXO1/LXRbeta complex, FOXA2, and LXRalpha on the hepatic enhancer of the apoA-I gene. PMID: 27404023
  7. Expression of certain LXR-dependent genes involved in cholesterol trafficking correlates with breast tumor characteristics but not time to recurrence. PMID: 28982861
  8. A positive association between placental PPARgamma mRNA levels and placental DHA levels with baby weight has been observed. PMID: 27578106
  9. GW3965 significantly increases the expression of liver X nuclear receptor beta (LXRbeta) mRNA, while the liver X nuclear receptor alpha (LXRalpha) mRNA expression remains relatively unchanged, and sensitizes gefitinib by inhibiting NF-kappa B (NF-kappaB) activation. PMID: 28178657
  10. The effects of LXR agonists on interleukin-8 (IL-8) secretion and nuclear factor-kappa B (NF-kappaB) activation in human umbilical vein endothelial cells (HUVECs) have been reported. PMID: 27489081
  11. Interactions among SNPs in nucleotide excision repair (NER) genes have been identified. PMID: 28115302
  12. Joint deletion of two short conserved motifs that bind UNR and DDX6 relieves repression of 4E-T-bound mRNA, partly reliant on the 4E-T-DDX6-CNOT1 axis. PMID: 27342281
  13. These results suggest that 25-HC promoted ADC cell migration and invasion in an LXR-dependent manner in the monoculture system, while in the coculture system, the 25-HC-induced IL-1beta secretion enhanced the effect of 25-HC in an LXR-independent manner. PMID: 28167281
  14. Protein level of LXRbeta protein was significantly reduced in focal cortical dysplasia. PMID: 27437943
  15. Distinct gene regulatory programs define the inhibitory effects of liver X receptors, NR1H2/NR1H3 and PPARG on cancer cell proliferation. PMID: 27401066
  16. These data describe a novel role for Unr in regulating cellular gene expression. PMID: 26936655
  17. Predominant cytoplasmic localization of LXRbeta, which occurs in colon cancer cells but not in normal colon epithelial cells, enabled LXR ligand-induced pyroptosis. PMID: 26450852
  18. These data identify a new mechanism of LXR regulation involving TIPARP, ADP-ribosylation and MACROD1. PMID: 26814197
  19. Intestinal activation of LXR reduces the production of chylomicrons by a mechanism dependent on the apical localization of SR-B1. PMID: 26602218
  20. The release of NER components such as DNA damage binding protein 2 (DDB2) and Xeroderma Pigmentosum complementation group C protein (XPC) following oxidative stress might potentially involve their apoptotic role rather than DNA repair function. PMID: 26263968
  21. LXRalpha expression was not altered in NAFLD. PMID: 26019035
  22. Recent high-throughput analyses of RNA-protein interactions indicate that Unr binds to a large subset of cellular mRNAs, suggesting that Unr may play a broader role in translational responses to cellular signals than previously believed. PMID: 26009170
  23. Destabilization of the torsioned conformation of a ligand side chain inverts the LXRbeta activity. PMID: 26434697
  24. LXRb is the dominant isoform in the rat myocardium, and the expression of both LXR isoforms (LXRa and LXRb) did not change after administration of T0901317. PMID: 25659329
  25. This study provides the first evidence showing that LXR activation reduces cadmium-induced apoptotic cell death of human renal proximal tubular cells by inhibiting reactive oxygen species production and JNK activation. PMID: 25980575
  26. Activation of LXRs interfered with the release of interleukin-6 from macrophages, thus inhibiting fibroblast activation and collagen release. PMID: 24618263
  27. LXR-b, through pannexin 1 interaction, can specifically induce caspase-1-dependent colon cancer cell death by pyroptosis. PMID: 25124554
  28. Data show that phospholipase C epsilon 1 (PLCE1) and liver X receptor-beta (LXR-beta) network interactions are significant contributory factors for genetic predisposition in gallbladder cancer. PMID: 24863943
  29. Studies indicate that no liver X receptor (LXR) modulator has successfully progressed beyond phase I clinical trials. PMID: 24832115
  30. A significant reduction in LXR-beta transcript has been reported in the testes of men with azoospermia. PMID: 24842676
  31. Structural analysis of the human retinoid X receptor alpha-liver X receptor beta (RXRalpha-LXRbeta) heterodimer on its cognate element, an AGGTCA direct repeat spaced by 4 nt, has been conducted. PMID: 24561505
  32. Treatment of human THP-1 macrophages with endogenous or synthetic LXR ligands stimulates both transcriptional and posttranscriptional pathways that result in the selective recruitment of the LXRalpha subtype to LXR-regulated promoters. PMID: 23686114
  33. LXR-beta plays roles in regulating endothelial cellular senescence, related to its antiatherogenic properties. PMID: 24398515
  34. Data indicate that LXR-beta genotypes (rs35463555) and (rs2695121) were associated with the risk of gallbladder cancer (GBC) compared to healthy controls, whereas LXR-alpha (rs7120118) was not associated with GBC risk. PMID: 23838803
  35. Variants in LXRalpha and LXRbeta genes are not potential contributors to the risk of metabolic syndrome and related traits in an Iranian population. PMID: 24100084
  36. Activation of LXR-alpha and LXR-beta suppresses the proliferation of human colon cancer cells. PMID: 22610535
  37. LXRbeta exhibits a nonnuclear function in endothelial cell caveolae/lipid rafts that involves crosstalk with estrogen receptor alpha. PMID: 23867501
  38. Molecular modeling of the interaction of 17(20)Z- and 17(20)E-pregna-5,17(20)-dien-21-oyl amides with the nuclear receptor LXRbeta has been performed. PMID: 23987069
  39. In human primary melanocytes, MNT-1, and B16 melanoma cells, TO901317, a synthetic LXR ligand, inhibited melanogenesis. PMID: 23223141
  40. Both LXRalpha and LXRbeta are expressed in rheumatoid arthritis fibroblast-like synoviocytes during inflammatory responses. FLS PMID: 22990668
  41. Activation of LXR reduced the binding of the transcriptional factors AP-1 and NF-kappaB to the ET-1 gene promoter region, thereby regulating gene expression. PMID: 23018104
  42. This article discusses the role of LXR in orchestrating lipid homeostasis and neuroinflammation in the brain. The ability of LXR to attenuate Alzheimer disease pathology makes them potential therapeutic targets for this neurodegenerative disease. [Review Article] PMID: 22766509
  43. Pharmacological activation of endothelial LXRs reduces angiogenesis by restraining cholesterol-dependent vascular endothelial growth factor receptor-2 compartmentation and signaling. PMID: 22723445
  44. Liver X receptors are activated by phospholipase A2 modified low density lipoproteins in human macrophages. PMID: 22367754
  45. The results of this study suggest that genetic variations in MMEL1, ECE1, ECE2, AGER, PLG, PLAT, NR1H3, MMP3, LRP1, TTR, NR1H2, and MMP9 genes do not play a major role among the Finnish AD patient cohort. PMID: 22027013
  46. LXR is involved in the metabolism and inflammation in human diseases; nonalcoholic fatty liver disease (NAFLD) is classically associated with lipid metabolic disorders and inflammatory responses. PMID: 22257474
  47. RXRalpha and LXR activate two promoters in placenta- and tumor-specific expression of PLAC1. PMID: 21937108
  48. Concomitant activation of ERbeta and inhibition of LXRbeta prevents 27-hydroxycholesterol effects and reduces the progression of Parkinson's disease by precluding tyrosine hydroxylase reduction and alpha-synuclein accumulation. PMID: 21951066
  49. There was no association between NR1H3 SNPs and pre-eclampsia, but the NR1H2 polymorphism rs2695121 was strongly associated with preeclampsia. PMID: 22029530
  50. We concluded that the LXR-alpha/beta gene expression ratio is a critical factor in activating POMC gene expression in ACTH-secreting pituitary adenomas. PMID: 21356276

Show More

Hide All

Database Links

HGNC: 7965

OMIM: 600380

KEGG: hsa:7376

STRING: 9606.ENSP00000253727

UniGene: Hs.432976

Protein Families
Nuclear hormone receptor family, NR1 subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Ubiquitous.

Q&A

What is NR1H2 and why is it an important research target?

NR1H2 (Nuclear Receptor Subfamily 1, Group H, Member 2), also known as Liver X Receptor beta (LXR-β), belongs to the nuclear receptor superfamily. It functions as a key regulator of macrophage activity, controlling transcriptional programs involved in lipid homeostasis and inflammation. NR1H2 preferentially binds to double-stranded oligonucleotide direct repeats with the consensus half-site sequence 5'-AGGTCA-3' and 4-nucleotide spacing (DR-4) . It plays a critical role in regulating cholesterol uptake through MYLIP-dependent ubiquitination of LDLR, VLDLR, and LRP8 . These physiological functions make NR1H2 a significant target in research related to metabolic disorders, inflammation, and lipid regulation.

Which applications are most commonly validated for NR1H2 antibodies?

Based on comprehensive antibody validation studies, NR1H2 antibodies have been successfully employed in multiple experimental applications:

ApplicationFrequency in LiteratureNotes
Western Blot (WB)High (12+ publications)Most widely validated application
Immunohistochemistry (IHC)ModerateValidated across multiple tissue types
ELISAModerateConsistently reported in antibody specifications
ChIP/ChIP-seqLow (1+ publications)Emerging application for studying DNA binding
Immunofluorescence (IF)ModerateParticularly useful for subcellular localization studies

For optimal results, researchers should select antibodies specifically validated for their intended application, as reactivity and optimal dilution can vary significantly between techniques .

What dilution ranges are recommended for Western blot applications with NR1H2 antibodies?

Western blot (WB) protocol optimization is critical for detecting NR1H2. Based on multiple manufacturer specifications, the following dilution ranges are recommended:

Antibody TypeRecommended WB DilutionSource
Mouse Monoclonal1:500-1:2000Proteintech
Mouse Monoclonal1:500-1:5000NovoPro
Rabbit PolyclonalVariable (application-dependent)Multiple sources

It is strongly recommended to perform preliminary titration experiments with your specific sample types, as optimal dilutions may vary depending on sample source, protein expression levels, and detection methods . The observed molecular weight for NR1H2 in Western blot applications is consistently reported at approximately 51 kDa .

How do epitope selection and antibody clonality affect NR1H2 detection across different experimental contexts?

Epitope specificity significantly impacts experimental outcomes when investigating NR1H2. The available antibody products target distinct regions of the protein:

Target RegionAntibody ClonalityNotable CharacteristicsRecommended Applications
N-TerminalPolyclonalRecognizes amino acids 1-50, often includes binding to GNGPPQPGAP sequence WB, IF, FACS
Internal (AA 150-250)PolyclonalContains the KEAGMREQCV LSEEQIRKKK sequence WB, IF
Full-length (AA 1-461)MonoclonalRecognizes complete protein structureWB, ELISA, IF

Monoclonal antibodies offer higher specificity but potentially lower sensitivity, making them ideal for applications requiring precise target recognition. Polyclonal antibodies typically provide stronger signals by recognizing multiple epitopes but may exhibit higher background in some applications .

For cross-species studies, researchers should note that sequence homology varies by region. Antibodies targeting highly conserved domains show greater cross-reactivity (human/mouse/rat/pig) , while those targeting variable regions may be species-specific.

What are the optimal tissue fixation and antigen retrieval methods for immunohistochemical detection of NR1H2?

Immunohistochemical detection of NR1H2 requires careful consideration of tissue preparation methodology:

Based on systematic studies across multiple tissue types (n=1267 images analyzed), successful IHC protocols for NR1H2 typically employ:

  • Fixation: 10% neutral buffered formalin fixation for 24-48 hours yields optimal results

  • Sectioning: 4-5μm sections mounted on positively charged slides

  • Antigen Retrieval: Heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0) for 20 minutes

  • Blocking: 3-5% normal serum (species-dependent on secondary antibody)

  • Primary Antibody Incubation: Dilution ranges of 1:20-1:50 for optimal staining

  • Detection System: HRP-polymer based detection systems yield superior results compared to biotin-based methods

The Human Protein Atlas project has comprehensively validated NR1H2 antibodies across 44 normal human tissues and 20 common cancer types, providing a robust reference for expected staining patterns .

How should researchers address potential cross-reactivity concerns when using NR1H2 antibodies?

Cross-reactivity validation is essential for meaningful NR1H2 research. Multiple approaches should be employed:

  • Knockout/Knockdown Controls: The definitive validation control utilizes samples from NR1H2 knockout/knockdown models. Published literature includes at least 4 studies using this gold-standard approach .

  • Peptide Competition Assays: Pre-incubation with immunizing peptide should abolish specific staining. Some manufacturers offer blocking peptides specifically designed for their NR1H2 antibodies .

  • Cross-Species Reactivity Analysis: Predicted reactivity based on epitope conservation:

    • Human: 100% (reference sequence)

    • Dog: 90% homology

    • Guinea Pig: 86% homology

    • Rat: 86% homology

    • Horse: 79% homology

  • Multiple Antibody Comparison: Using antibodies targeting different epitopes can confirm specificity. The Human Protein Atlas employed this strategy across their tissue microarray validation studies .

When publishing results, researchers should document which validation approaches were employed, as these significantly impact the interpretation of experimental outcomes.

What is the normal tissue distribution pattern of NR1H2 and how should this influence experimental design?

Understanding NR1H2's tissue-specific expression pattern is crucial for experimental design and interpretation:

Tissue TypeExpression LevelNotable Considerations
LiverHighPrimary tissue for antibody validation; consistent detection across studies
AdrenalModerateExpression varies by adrenal region
Brain RegionsVariableExpression differs across anatomical regions (amygdala, hippocampus, etc.)
LungModerateExpression changes in pathological states (emphysema, carcinoma)
Lymphoid TissuesModerateDetected in lymph nodes and spleen
Reproductive TissuesVariableDetected in testis, ovary, prostate, and uterus

The comprehensive Human Protein Atlas evaluation analyzed NR1H2 expression across 44 normal tissues, 11 brain regions, and 25 disease tissues, establishing baseline expression patterns for comparative studies . When designing experiments targeting specific tissues, researchers should select antibodies validated in their tissue of interest, as detection efficiency can vary by tissue type.

What methodological considerations are important for studying NR1H2 in disease models?

When investigating NR1H2 in pathological contexts, several methodological considerations are critical:

  • Disease-Specific Expression Changes: The Human Protein Atlas has documented NR1H2 expression changes in multiple disease states, including:

    • Alzheimer's disease (brain tissue)

    • Atherosclerosis (arterial tissue)

    • Carcinomas (lung, breast, colon, ovary, pancreas, prostate)

    • Inflammatory conditions (Crohn's disease, rheumatoid arthritis)

  • Control Selection: Appropriate control tissues must be matched for:

    • Age and sex of donor

    • Anatomical region (particularly critical for brain studies)

    • Fixation methodology and duration

    • Processing method

  • Quantification Approaches: For comparing disease vs. normal tissues:

    • Use digital image analysis with standardized exposure settings

    • Employ multiple antibodies targeting different epitopes

    • Include isotype controls to assess background staining

    • Consider multiplexed approaches to evaluate co-localization with disease markers

  • Statistical Analysis: When comparing expression levels between disease models and controls, apply appropriate statistical tests based on sample distribution and experimental design.

What are the technical challenges in ChIP applications with NR1H2 antibodies and how can they be addressed?

Chromatin Immunoprecipitation (ChIP) with NR1H2 antibodies presents specific technical challenges:

  • Cross-linking Optimization: As a nuclear receptor that binds DNA, NR1H2 requires careful cross-linking optimization:

    • Standard formaldehyde cross-linking (1% for 10 minutes) may be insufficient

    • Consider dual cross-linking approaches (DSG followed by formaldehyde)

    • Evaluate cross-linking efficiency through sonication testing

  • Antibody Selection: Only specific NR1H2 antibodies are validated for ChIP applications:

    • Look for antibodies specifically tested in ChIP/ChIP-seq applications

    • Consider antibodies targeting the DNA-binding domain for direct binding studies

    • Polyclonal antibodies targeting internal regions have shown success in published ChIP applications

  • Control Strategies:

    • Input controls are essential for normalization

    • IgG controls (matched to host species) assess non-specific binding

    • Known NR1H2 binding sites can serve as positive controls

    • Regions without predicted binding sites serve as negative controls

  • Data Analysis Considerations:

    • NR1H2 binds preferentially to DR-4 motifs (direct repeats with consensus half-site sequence 5'-AGGTCA-3')

    • Peak calling algorithms should be optimized for transcription factor binding patterns

    • Integration with transcriptomic data enhances functional interpretation

How can multiplexed immunofluorescence approaches be optimized for studying NR1H2 interactions with other proteins?

Multiplexed immunofluorescence offers powerful insights into NR1H2's protein-protein interactions and cellular context:

  • Antibody Compatibility Planning:

    • Select NR1H2 antibodies from different host species than interaction partner antibodies

    • If using same-species antibodies, employ sequential staining with intermediate blocking steps

    • Test for cross-reactivity between secondary antibodies

  • Epitope Retrieval Harmonization:

    • Identify compatible antigen retrieval conditions for all target proteins

    • Consider tyramide signal amplification for low-abundance targets

    • Validate multiplex protocol against single-stain controls

  • Spectral Considerations:

    • Select fluorophores with minimal spectral overlap

    • Include single-stain controls for spectral unmixing

    • Consider autofluorescence quenching (especially important in liver tissue)

  • Analysis Approaches:

    • Employ cell-by-cell colocalization analysis

    • Consider proximity ligation assays for direct interaction studies

    • Quantify subcellular distribution patterns using digital image analysis

What experimental approaches can address contradictory findings in NR1H2 research?

Research discrepancies in NR1H2 studies often stem from methodological differences. Systematic approach to resolving contradictions includes:

  • Antibody Characterization Discrepancies: Different antibodies may yield contradictory results due to:

    • Epitope differences (N-terminal vs. internal vs. C-terminal)

    • Clonality differences (monoclonal vs. polyclonal)

    • Cross-reactivity with related proteins (particularly NR1H3/LXRα)

    Resolution Strategy: Employ multiple antibodies targeting different epitopes, including at least one validated through knockout/knockdown studies .

  • Species Differences: NR1H2 function may vary between species:

    • Human NR1H2: 461 amino acids

    • Mouse/Rat: Sequence divergence in regulatory domains

    • Other species: Variable conservation

    Resolution Strategy: Confirm antibody reactivity in your species of interest through validated controls.

  • Isoform Detection: Alternative splicing may generate multiple NR1H2 isoforms:

    • Standard Western blot protocols may not resolve all isoforms

    • Isoform distribution may vary by tissue type

    Resolution Strategy: Use high-resolution gel systems (gradient gels) and antibodies targeting conserved regions.

  • Context-Dependent Regulation: NR1H2 function is modulated by:

    • Ligand binding state

    • Post-translational modifications

    • Cofactor availability

    Resolution Strategy: Characterize experimental conditions thoroughly, including cell type, treatment conditions, and timepoints.

What are the best practices for quantifying NR1H2 expression levels across different experimental platforms?

Quantitative assessment of NR1H2 requires platform-specific considerations:

  • Western Blot Quantification:

    • Use housekeeping controls appropriate to your experimental context

    • Consider total protein normalization (Ponceau S, REVERT staining)

    • Ensure signal is within linear range of detection

    • Report relative expression with appropriate statistical analysis

  • Immunohistochemistry Quantification:

    • Standardize staining conditions across all samples

    • Use digital image analysis for objective quantification

    • Report both staining intensity and percentage of positive cells

    • Consider established scoring systems (H-score, Allred score)

  • qPCR for mRNA Expression:

    • Validate primer specificity through melt curve analysis and sequencing

    • Use multiple reference genes for normalization

    • Consider absolute quantification with standard curves

    • Report fold-changes with appropriate statistical analysis

  • Cross-Platform Validation:

    • Correlate protein levels (Western blot) with mRNA expression (qPCR)

    • Validate spatial distribution with immunohistochemistry

    • Consider functional assays to correlate expression with activity

These methodological approaches ensure robust, reproducible quantification across experimental platforms.

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.