NFIX is a member of the nuclear factor I (NFI) family, characterized by its ability to bind the DNA sequence 5'-TTGGCNNNNNGCCAA-3' in promoters and regulate transcription . Key roles include:
Developmental regulation: Critical in muscle and central nervous system development .
Cancer biology: Promotes glioblastoma (GBM) cell migration via Ezrin upregulation and exhibits context-dependent oncogenic or tumor-suppressive roles in other cancers .
Isoforms: Six isoforms reported in humans, with a canonical length of 502 amino acids and a mass of 55.1 kDa .
Glioblastoma: NFIX antibody identified NFIX-Ezrin axis-driven migration in GBM cells, with knockdown studies showing reduced tumor growth in mice .
Prostate and Breast Cancer: Detected NFIX overexpression linked to SOX4 and acyl-CoA synthetase 4 pathways .
Hippocampal Studies: Used in rodent brain tissue to localize NFIX in excitatory and inhibitory neurons .
NFIX antibodies are rigorously validated:
Western Blot: Detects bands at ~45–55 kDa in brain tissue (human, mouse, rat) .
IHC-Paraffin: Optimized antigen retrieval with citrate buffer (pH 6.0) or TE buffer (pH 9.0) .
Species Cross-Reactivity: Broad reactivity across mammals, including pig, dog, and zebrafish .
NFIX (Nuclear Factor I/X) is a sequence-specific transcription factor belonging to the Nuclear Factor I family. It functions as a CCAAT-binding transcription factor and is also known by several aliases including CTF, NF1A, NF1-X, MRSHSS, NF-I/X, and SOTOS2 . The NFIX protein (Nuclear factor 1 X-type) plays crucial roles in regulating gene expression during development and in various cellular processes.
Research has shown that NFIX is particularly important in hematopoietic stem and progenitor cells (HSPCs), where it promotes ex vivo growth, cytokine hypersensitivity, and survival of primitive hematopoietic populations . NFIX has been identified as a transcriptional regulator of c-Mpl (the thrombopoietin receptor), revealing one molecular pathway through which NFIX influences HSPCs . This regulation appears to be direct, as chromatin immunoprecipitation studies have confirmed NFIX binding to the c-Mpl promoter region .
Researchers can access several types of NFIX antibodies optimized for different experimental applications:
Polyclonal antibodies: These are commonly generated in rabbits using synthetic peptides directed toward specific regions of human NFIX, such as the middle region . Polyclonal antibodies recognize multiple epitopes on the NFIX protein, potentially providing stronger signals in applications like Western blotting.
Region-specific antibodies: These target defined regions of the NFIX protein:
Application-optimized antibodies: NFIX antibodies validated specifically for:
When selecting an NFIX antibody, researchers should consider the specific region of interest and ensure the antibody has been validated for their intended application.
Proper storage and handling of NFIX antibodies are critical for maintaining their activity and specificity. Based on manufacturer recommendations:
Long-term storage: Store at -20°C in small aliquots to prevent freeze-thaw cycles . Repeated freeze-thaw cycles can degrade antibody quality and reduce binding efficiency.
Working dilutions: Prepare fresh working dilutions on the day of the experiment whenever possible.
Buffer considerations: Many NFIX antibodies are supplied in 1x PBS buffer with 0.09% (w/v) sodium azide and 2% sucrose . The sodium azide serves as a preservative, while sucrose helps maintain protein stability during freeze-thaw cycles.
Handling precautions: Avoid contamination, and handle according to good laboratory practices, particularly since sodium azide is toxic.
Following these storage and handling recommendations will help ensure consistent and reliable results when using NFIX antibodies in research protocols.
Optimizing Western blotting with NFIX antibodies requires careful attention to several parameters:
Sample preparation:
Use fresh tissue/cell lysates whenever possible
Include protease inhibitors during lysis to prevent NFIX degradation
Normalize protein loading (30-50 μg total protein per lane is typically suitable)
Antibody dilution and incubation:
Start with the manufacturer's recommended dilution (typically 1:500 to 1:2000)
Perform a dilution series to determine optimal concentration for your specific sample
Incubate primary antibody overnight at 4°C for optimal binding
Blocking and washing:
Use 5% non-fat dry milk or BSA in TBST for blocking
Perform at least 3-5 washes (5-10 minutes each) with TBST after both primary and secondary antibody incubations
Controls:
Detection:
The expected molecular weight of NFIX is approximately 55-60 kDa, though isoforms and post-translational modifications can result in multiple bands.
When using NFIX antibodies for immunohistochemistry, consider these methodological best practices:
Tissue preparation:
Fixation: 10% neutral-buffered formalin is typically suitable
Section thickness: 4-6 μm is optimal for most applications
Deparaffinization and rehydration should be complete to ensure antibody access
Antigen retrieval:
Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Pressure cooker or microwave methods may provide better results than water bath methods
Blocking:
Block endogenous peroxidase activity with 0.3-3% hydrogen peroxide
Use normal serum (from the same species as the secondary antibody) to reduce background
Antibody incubation:
Dilute according to manufacturer's recommendations (usually 1:100 to 1:500)
Incubate at 4°C overnight for optimal staining
Consider using a humidity chamber to prevent section drying
Detection systems:
Polymer-based detection systems often provide better sensitivity with less background
Counterstain with hematoxylin to visualize tissue architecture
Use mounting media appropriate for your detection system (aqueous for fluorescent detection)
Controls:
Include positive control tissues known to express NFIX
Include a negative control by omitting primary antibody
Consider using tissues from NFIX knockout models as specificity controls if available
Remember that optimal conditions may vary based on the specific NFIX antibody and tissue type being analyzed.
Validating antibody specificity is crucial for generating reliable research data. For NFIX antibodies, consider these validation approaches:
Molecular validation:
Immunogen sequence analysis:
Species cross-reactivity testing:
Orthogonal validation:
Compare results with a second NFIX antibody targeting a different epitope
Correlate protein detection with mRNA levels using RT-PCR or RNA-seq
Consider using multiple applications (WB, IHC, IF) to confirm consistent detection patterns
Functional validation:
Thorough validation ensures that your experimental observations truly reflect NFIX biology rather than antibody artifacts.
NFIX antibodies can provide valuable insights into transcriptional regulation mechanisms through several advanced experimental approaches:
Chromatin Immunoprecipitation (ChIP):
Use NFIX antibodies to immunoprecipitate chromatin fragments bound to NFIX
Research has demonstrated NFIX binding to the c-Mpl promoter region
Protocol considerations:
Cross-link cells with 1% formaldehyde
Sonicate chromatin to 200-500bp fragments
Immunoprecipitate with NFIX antibody (2-5 μg per reaction)
Analyze bound regions by qPCR or sequencing (ChIP-seq)
Co-Immunoprecipitation (Co-IP):
Identify protein interaction partners of NFIX
Use NFIX antibodies to pull down NFIX and associated proteins
Analyze by mass spectrometry or Western blotting with antibodies against suspected interacting partners
Electrophoretic Mobility Shift Assay (EMSA):
Study direct DNA binding of NFIX to target sequences
Use NFIX antibodies in supershift assays to confirm specificity
Include putative binding sites such as those identified in the c-Mpl promoter region
Promoter-Reporter Assays:
This multi-faceted approach can reveal not only which genes NFIX regulates but also the specific binding sites and mechanisms of regulation.
NFIX has emerged as an important regulator of hematopoietic stem and progenitor cells (HSPCs). Research has shown that NFIX promotes ex vivo growth, cytokine hypersensitivity, and survival of primitive hematopoietic populations . Investigating this role requires specialized methodologies:
Ex vivo culture systems:
Flow cytometry analysis:
Functional assays:
Colony-forming unit (CFU) assays to assess progenitor function
Competitive repopulation assays to evaluate in vivo stem cell activity
Cytokine response studies, particularly with TPO/c-MPL signaling pathway
Molecular mechanism investigations:
| Parameter | Control Cells | NFIX+ Cells | Significance |
|---|---|---|---|
| c-MPL transcript expression | Baseline | 2-fold increase | p = 0.028 |
| c-MPL cell surface protein | Baseline | 2-fold increase | p = 0.042 |
| Cell growth in reduced cytokines | Significantly reduced | Maintained | p = 0.048 |
| Apoptosis in reduced cytokines | Increased | Unaffected | p = 0.032 |
These methodologies can help researchers elucidate the mechanisms through which NFIX regulates hematopoietic stem cell function and survival.
The regulatory relationship between NFIX and c-Mpl (thrombopoietin receptor) represents an important molecular pathway in hematopoietic research. To investigate this relationship, consider these approaches:
Gene expression analysis:
Protein expression analysis:
Promoter analysis and direct binding:
Functional relationship studies:
Downstream signaling analysis:
Evaluate activation of pathways downstream of c-MPL (JAK/STAT, MAPK, PI3K/AKT)
Compare phosphorylation status of key signaling molecules
Determine how NFIX overexpression affects response to TPO stimulation
These methodological approaches can provide a comprehensive understanding of how NFIX regulates c-Mpl expression and the functional consequences of this regulation in hematopoietic cells.
High background or non-specific binding can compromise experimental results. Consider these methodological solutions to common problems:
Antibody-related factors:
Dilution issues: Try a more dilute antibody solution (1:1000 instead of 1:500)
Quality control: Check antibody purity (affinity-purified antibodies like those in search result typically give cleaner results)
Consider using a different NFIX antibody targeting another epitope region
Use freshly prepared working solutions of antibody
Sample preparation issues:
Overfixation: Reduce fixation time or concentration
Incomplete blocking: Increase blocking time or concentration
Autofluorescence: Include a quenching step or use a different detection system
Cross-reactivity with endogenous biotin: Use a biotin-free detection system
Protocol optimization:
More stringent washing: Increase wash duration, number, or detergent concentration
Better blocking: Try different blocking agents (BSA, normal serum, commercial blockers)
Reduce secondary antibody concentration
Include 0.1-0.3% Triton X-100 in antibody diluent to reduce non-specific membrane binding
Validation steps:
Detection system considerations:
High sensitivity detection systems may amplify background
Consider using polymer-based detection instead of avidin-biotin systems
Reduce substrate development time
Systematic troubleshooting of these factors can help achieve clean, specific NFIX detection.
Detecting low NFIX expression levels requires careful optimization of antibody concentration and detection methods:
Antibody titration:
Perform a dilution series (e.g., 1:100, 1:250, 1:500, 1:1000, 1:2000)
Use a positive control sample with known NFIX expression
Select the dilution that provides the best signal-to-noise ratio, not necessarily the strongest signal
Consider using concentrated antibody preparations (e.g., 0.5 mg/ml concentration)
Sample enrichment techniques:
Immunoprecipitation before Western blotting
Cell sorting to enrich for populations with higher NFIX expression
Nuclear extraction (since NFIX is a nuclear protein)
Concentration of dilute samples
Signal amplification methods:
For Western blotting:
Use high-sensitivity ECL substrates
Consider longer exposure times
Try fluorescent secondary antibodies with digital imaging
For IHC/ICC:
Use tyramide signal amplification (TSA)
Try polymer-based detection systems
Consider biotin-streptavidin amplification (if endogenous biotin is not an issue)
Experimental controls:
Include a dilution series of recombinant NFIX protein to establish detection limits
Use samples with NFIX overexpression as positive controls
Compare results with a second NFIX antibody targeting a different epitope
Optimization based on sample type:
Fresh vs. frozen tissue may require different antibody concentrations
Cell lines vs. primary cells may have different optimal conditions
Consider specialized fixation protocols for low-abundance nuclear proteins
Through methodical optimization, you can establish conditions that enable detection of even low levels of NFIX expression while maintaining specificity.
Proper controls are essential for generating reliable and interpretable data when studying NFIX. Include these controls in your experimental design:
Antibody validation controls:
Expression controls:
Positive control: Include samples known to express NFIX (based on literature)
Negative control: Use samples where NFIX expression is absent or knocked down
Overexpression control: Samples with forced NFIX expression serve as positive controls and help identify antibody specificity
Functional controls:
Technical controls:
Loading controls for Western blot (β-actin, GAPDH, histone H3 for nuclear extracts)
Housekeeping genes for RT-qPCR (GAPDH, β-actin, 18S rRNA)
Staining controls for flow cytometry (FMO - Fluorescence Minus One)
Experimental design controls:
Time course experiments to capture dynamic changes
Dose-response relationships for treatments
Biological replicates (different donors/animals) and technical replicates
Detecting NFIX in specific cell populations like hematopoietic stem cells (HSCs) requires specialized approaches:
Cell isolation and enrichment:
For HSCs, use standard markers (LSK: Lin-Sca-1+c-Kit+) for flow cytometry sorting
Consider magnetic bead separation for pre-enrichment
Culture cells under conditions that maintain stemness (e.g., with appropriate cytokines)
Note that extended culture may affect immunophenotype, as NFIX+ LSK cells show accelerated loss of the LSK phenotype
Detection method selection:
For rare populations like HSCs:
Flow cytometry provides single-cell resolution
Immunofluorescence allows visualization of subcellular localization
Western blotting may require larger cell numbers (pooled samples)
Consider multiparameter analysis combining NFIX with stem cell markers
Protocol modifications:
Gentle fixation to preserve HSC markers and NFIX epitopes
Optimize permeabilization for nuclear antigen detection
Use low-background detection systems
For flow cytometry, include dead cell discrimination dyes
Functional correlation:
Single-cell approaches:
Consider single-cell protein detection methods (CyTOF, CITE-seq)
Correlate protein expression with single-cell transcriptomics
Image-based single-cell analysis (imaging flow cytometry)
Research has shown that NFIX+ HSPCs display distinct properties, including resistance to cytokine deprivation and reduced apoptosis during ex vivo culture . These functional characteristics can help confirm successful detection of NFIX-expressing stem cell populations.
NFIX research is providing important insights into stem cell biology and disease mechanisms:
Hematopoietic stem cell regulation:
Transcriptional networks:
Cell survival mechanisms:
Differentiation pathways:
Disease implications:
Continued research on NFIX will likely expand our understanding of normal stem cell regulation and provide insights into disease mechanisms and potential therapeutic approaches.
Emerging technologies promise to advance NFIX research in several key areas:
Advanced genomic approaches:
CUT&RUN or CUT&Tag for more precise mapping of NFIX binding sites
HiChIP or Micro-C to identify long-range chromatin interactions mediated by NFIX
CRISPR screening to identify functional partners or downstream effectors
Base editing or prime editing for precise genetic manipulation
Single-cell technologies:
Single-cell multiomics (RNA-seq + ATAC-seq + protein detection)
Spatial transcriptomics to understand NFIX function in tissue context
Live cell imaging of NFIX dynamics using engineered reporter systems
Single-cell proteomics to detect low-abundance NFIX in specific cell populations
Structural biology:
Cryo-EM of NFIX-DNA complexes
Hydrogen-deuterium exchange mass spectrometry to map binding interfaces
AlphaFold or other AI-based structural prediction tools for NFIX interactions
Computational approaches:
Machine learning to predict NFIX binding sites and functional outcomes
Network analysis to position NFIX within broader regulatory networks
Integration of multi-omic datasets to build comprehensive models of NFIX function
Advanced antibody technologies:
Recombinant antibodies with enhanced specificity
Nanobodies for improved access to nuclear antigens
Proximity labeling strategies using NFIX antibodies to identify interaction partners
These emerging technologies will provide deeper insights into NFIX biology and potentially reveal new therapeutic approaches targeting NFIX-regulated pathways.