The table below lists top commercial antibodies validated for NELFCD detection:
These antibodies target epitopes in the C-terminal region and are used for Western blotting (WB), immunohistochemistry (IHC), and chromatin immunoprecipitation (ChIP) .
Overexpression: NELFCD mRNA and protein levels are elevated in 85% of CRC tumors compared to adjacent normal tissues (P < 0.05) .
Functional Impact:
Clinical Relevance: High NELFCD correlates with advanced tumor stages and metastasis .
Genetic Link: The rs163800 SNP near NELFCD/CTSZ loci increases jaundice-stage progression risk in PBC (OR = 2.16, P = 8.57 × 10⁻⁸) .
Mechanism: Risk alleles elevate NELFCD expression while suppressing CTSZ, dysregulating MAPK and hormonal signaling pathways .
NELFCD antibodies enable researchers to:
NELFCD (Negative Elongation Factor C/D, also known as TH1L protein) functions as an essential component of the NELF complex, which negatively regulates transcription elongation by RNA polymerase II. The protein acts through association with the DSIF complex to cause transcriptional pausing, a mechanism counteracted by the P-TEFb kinase complex. Additionally, the NELF complex appears to be involved in HIV-1 latency, possibly through the recruitment of PCF11 to paused RNA polymerase II . Understanding NELFCD's function provides critical context for interpreting experimental results when using antibodies against this target.
NELFCD antibodies have been validated for several detection methods, with Western Blot (WB) being the most consistently reported application across suppliers. Some antibodies also demonstrate compatibility with immunohistochemistry (IHC), immunoprecipitation (IP), ELISA, and flow cytometry (FCM), depending on the specific product . When designing experiments, researchers should consider that rabbit polyclonal antibodies against NELFCD typically show reactivity with human and mouse samples at dilution ranges of 1:500-2000 for Western Blot applications .
NELFCD antibodies are typically formulated as liquids in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide . For optimal preservation of activity, these antibodies should be stored at -20°C for up to one year from the date of receipt. Researchers should take precautions to avoid repeated freeze-thaw cycles, as this can significantly degrade antibody performance over time . Aliquoting the antibody upon first thaw is recommended practice to minimize the impact of multiple freeze-thaw events on antibody integrity.
When performing Western blot analysis, researchers should expect to detect NELFCD at approximately 57.3 kilodaltons (kDa), which is the reported molecular mass for this protein . This information is crucial for accurate identification of bands during Western blot analysis and for distinguishing between specific binding and potential non-specific interactions that may appear at different molecular weights.
For functional studies investigating the role of NELFCD in cellular processes, RNA interference using short hairpin RNA (shRNA) has proven effective. As demonstrated in colorectal cancer research, shRNA constructs targeting NELFCD RNA sequences (e.g., 5′-GGAACAGATGATTGCACATAC-3′) can be transfected into cell lines such as SW480 and HCT-116 . After transfection, researchers should select infected cells expressing the vector using puromycin (5 μg/mL) for approximately ten consecutive days. The efficiency of NELFCD knockdown should be validated through both qRT-PCR and Western blotting to ensure significant reduction in expression before proceeding with functional assays .
Quantitative real-time PCR (qRT-PCR) has been established as an effective method for quantifying NELFCD expression in tissue samples. The protocol involves RNA extraction, cDNA synthesis, and amplification using NELFCD-specific primers. For normalization of RNA concentration, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) serves as an internal control. The 2−ΔΔCT method can be used to calculate the relative amount of NELFCD compared with GAPDH expression . The amplification protocol typically includes an initial denaturation step at 98°C for 2 minutes, followed by 40 cycles of 98°C for 10 seconds, 60°C for 10 seconds, and 68°C for 30 seconds, performed in instruments such as the Light Cycler 480 II .
Studies on colorectal cancer have revealed that NELFCD is significantly overexpressed in tumor tissues compared to adjacent non-tumor tissues (P<0.05) . Functional studies demonstrate that knockdown of NELFCD significantly impairs cancer cell proliferation, migration, and invasion abilities while facilitating apoptosis in vitro. Additionally, NELFCD knockdown inhibits tumorigenesis in vivo, suggesting its potential role as an oncogene . These findings indicate that NELFCD expression levels may have clinical relevance in cancer progression, and antibodies against NELFCD serve as valuable tools for investigating this correlation in different cancer types.
When selecting between monoclonal and polyclonal NELFCD antibodies, researchers should consider their specific experimental requirements. Polyclonal antibodies, such as rabbit anti-NELFCD, recognize multiple epitopes on the target protein, potentially offering higher sensitivity but with increased risk of cross-reactivity . These are advantageous for applications like Western blot and initial characterization studies. Conversely, monoclonal antibodies recognize a single epitope, providing higher specificity but potentially lower sensitivity, making them more suitable for targeted studies focusing on specific protein domains or for applications where cross-reactivity must be minimized. The experimental context, including the species being studied and the specific application, should guide this selection.
When encountering non-specific binding with NELFCD antibodies, several optimization strategies can be implemented. First, increasing the blocking concentration (e.g., from 3% to 5% BSA or milk) and extending blocking time can reduce background. Second, optimizing antibody dilution is crucial—testing a range of dilutions from 1:500 to 1:2000 for Western blot applications allows identification of the optimal concentration that balances signal strength with specificity . Third, increasing wash frequency and duration removes weakly bound antibodies. Additionally, pre-adsorption of the antibody with blocking proteins or non-relevant proteins from the same species as the sample can reduce cross-reactivity. For persistent issues, switching to more specific detection methods or alternative antibodies raised against different epitopes of NELFCD may resolve the problem.
Confirming the specificity of NELFCD antibody binding requires multiple validation approaches. First, researchers should perform positive and negative controls in parallel with experimental samples. Using NELFCD overexpression systems as positive controls and NELFCD knockdown samples as negative controls provides reliable reference points . Second, competing peptide assays, where the antibody is pre-incubated with the immunizing peptide before application to the sample, can confirm binding specificity—true signals should disappear in this test. Third, detecting the target protein at the expected molecular weight (approximately 57.3 kDa) in Western blot applications provides another layer of validation . Finally, cross-validating results using multiple antibodies targeting different epitopes of NELFCD or using alternative detection methods strengthens confidence in the observed results.
Multiple factors influence the reproducibility of NELFCD antibody-based experiments. First, antibody batch variability can impact results, particularly with polyclonal antibodies where different production lots may have varying epitope recognition profiles. Second, storage conditions significantly affect antibody stability—improper storage or repeated freeze-thaw cycles can degrade antibody quality . Third, protocol standardization, including consistent blocking, washing, and incubation times, reduces variability between experiments. Fourth, sample preparation methods, including protein extraction techniques and buffer compositions, influence antigen accessibility and preservation. Finally, the cellular or tissue context itself can affect results, as NELFCD expression and localization may vary with cell type, growth conditions, and treatment protocols. Maintaining detailed records of all experimental conditions and reagent sources is essential for troubleshooting reproducibility issues.
When comparing NELFCD expression across different samples or conditions, proper normalization is critical for valid interpretation. For qRT-PCR analysis, researchers should use established housekeeping genes such as GAPDH as internal controls. The 2−ΔΔCT method provides a standardized approach for calculating relative NELFCD expression using these reference genes . For protein-level analysis via Western blot, normalization to loading controls such as β-actin, GAPDH, or total protein (using techniques like Stain-Free gels) is essential. When analyzing immunohistochemistry data, researchers should employ established scoring systems that account for both staining intensity and the percentage of positive cells. Additionally, technical and biological replicates should be included in the experimental design, and statistical methods appropriate for the data distribution should be applied to determine significance.
The statistical approach for analyzing NELFCD expression differences depends on the experimental design and data characteristics. For comparing two groups (e.g., tumor vs. normal tissue), paired or unpaired t-tests are appropriate when data follow normal distribution . For non-normally distributed data, non-parametric tests such as Mann-Whitney or Wilcoxon signed-rank tests should be used. When comparing multiple groups, ANOVA (for parametric data) or Kruskal-Wallis tests (for non-parametric data) with appropriate post-hoc tests are recommended. For correlation analyses between NELFCD expression and clinical parameters, Pearson or Spearman correlation coefficients should be calculated based on data normality. In clinical studies, Kaplan-Meier survival analysis with log-rank tests can evaluate the relationship between NELFCD expression levels and patient outcomes. Multivariate analyses, such as Cox regression, can assess whether NELFCD is an independent prognostic factor while controlling for other variables.
Comparing results obtained with different NELFCD antibodies requires careful methodology. First, researchers should create a standardized reference sample set that can be probed with each antibody under investigation. Second, maintaining identical experimental conditions (sample preparation, protocols, detection methods) for all antibodies being compared minimizes procedural variability. Third, quantitative analysis using standardized methods (such as densitometry for Western blots or standardized scoring for immunohistochemistry) allows direct comparison of signal intensity and specificity. Fourth, statistical correlation analysis between results from different antibodies can quantify their agreement. Finally, validating observed patterns with orthogonal techniques (e.g., mass spectrometry or mRNA analysis) provides additional confirmation. When reporting comparative antibody results, researchers should clearly document the specific catalog numbers, clones, and experimental conditions to facilitate reproducibility.
NELFCD antibodies serve as powerful tools for investigating transcriptional dysregulation in cancer. Researchers can employ chromatin immunoprecipitation (ChIP) assays with NELFCD antibodies to identify genomic regions where the NELF complex is involved in transcriptional pausing, potentially revealing aberrant regulation in cancer cells. Studies have demonstrated that NELFCD is overexpressed in colorectal cancer tissues compared to adjacent normal tissues, with knockdown experiments showing significant impairment of cancer cell proliferation, migration, and invasion . This suggests NELFCD's potential role in promoting oncogenic processes. By combining NELFCD antibody-based detection with RNA sequencing data, researchers can correlate NELFCD binding with changes in gene expression patterns, providing insights into the mechanistic basis of cancer-specific transcriptional signatures. These approaches help elucidate how dysregulation of transcriptional elongation contributes to cancer development and progression.
The NELF complex, which includes NELFCD, is implicated in HIV-1 latency through its role in transcriptional pausing. This mechanism may involve the recruitment of PCF11 to paused RNA polymerase II . NELFCD antibodies can be instrumental in investigating this process through several approaches. Chromatin immunoprecipitation (ChIP) assays using NELFCD antibodies can identify binding patterns at the HIV-1 long terminal repeat (LTR) promoter region in latently infected cells. Immunoprecipitation with NELFCD antibodies followed by mass spectrometry or Western blotting can identify interaction partners specific to HIV-infected cells, potentially revealing novel regulatory mechanisms. Additionally, researchers can use these antibodies to monitor changes in NELFCD localization and complex formation during latency establishment and reactivation. These approaches contribute to a deeper understanding of HIV latency mechanisms and may inform the development of strategies to eliminate viral reservoirs by targeting transcriptional elongation factors.
NELFCD antibodies can significantly contribute to drug discovery efforts targeting transcriptional elongation processes. First, they can be employed in high-throughput screening assays to identify compounds that disrupt NELF complex formation or function, potentially through displacement of NELFCD from its binding partners. Second, antibodies can be used in cellular assays to monitor changes in NELFCD localization or post-translational modifications in response to candidate drugs, providing insights into compound mechanisms. Third, ChIP-seq with NELFCD antibodies before and after drug treatment can map genome-wide changes in NELF complex binding, helping characterize drug effects on transcriptional regulation. Fourth, combining NELFCD antibody-based assays with reporter systems for genes under NELF regulation creates functional readouts for drug efficacy. Finally, in cancer models where NELFCD is overexpressed, antibodies can monitor whether drug treatments normalize NELFCD levels or activity, potentially correlating with therapeutic efficacy.
Recent advances in computational approaches are significantly enhancing NELFCD antibody applications. Machine learning models, including LLM-style, diffusion-based, and graph-based models, are being developed to predict antibody binding affinities with increasing accuracy . These models generate log-likelihood scores that correlate strongly with experimentally measured binding affinities, providing a reliable metric for ranking antibody designs . For NELFCD research, these computational tools can help design more specific antibodies by predicting epitope-paratope interactions and optimizing binding properties. Additionally, structure prediction tools like ImmuneBuilder2 and IgFold can generate structural models of NELFCD-antibody complexes, aiding in understanding binding mechanisms and guiding antibody engineering efforts . As these computational methods continue to improve, they promise to accelerate antibody development and optimization while reducing the need for extensive experimental screening.
NELFCD antibodies hold significant potential for integration with emerging single-cell analysis technologies. In single-cell proteomics approaches, NELFCD antibodies conjugated to heavy metals can be used in mass cytometry (CyTOF) to quantify NELFCD levels in individual cells while simultaneously measuring dozens of other proteins, providing insights into heterogeneous cell populations. For spatial transcriptomics, combining NELFCD antibody-based immunofluorescence with in situ RNA sequencing can correlate NELFCD protein localization with gene expression patterns at single-cell resolution within tissue contexts. In microfluidic-based single-cell Western blotting, NELFCD antibodies can detect protein levels in individual cells, enabling correlation between NELFCD expression and cellular phenotypes. Furthermore, proximity ligation assays using NELFCD antibodies can identify protein-protein interactions in single cells, revealing cell-specific regulatory networks. These applications would be particularly valuable for understanding heterogeneity in NELFCD-dependent transcriptional regulation across different cell types within tumors or during development.
Implementing affinity-based ranking of NELFCD antibodies could substantially improve experimental reproducibility. Recent research has demonstrated that log-likelihood scores from generative models correlate well with experimentally measured binding affinities, suggesting these scores can serve as reliable metrics for ranking antibody designs . By systematically characterizing and ranking commercially available NELFCD antibodies based on affinity, specificity, and performance metrics, researchers could make more informed selections for their specific applications. This approach would involve generating a standardized dataset of binding affinities and cross-reactivity profiles for different NELFCD antibodies across various applications and sample types. The resulting ranking system would help researchers select antibodies with the optimal characteristics for their specific experimental needs, reducing variability between studies and enhancing reproducibility. Additionally, this systematic approach could identify gaps in the current antibody landscape, guiding the development of new antibodies with improved properties for understudied applications or epitopes.
Researchers beginning work with NELFCD antibodies should prioritize several critical considerations to ensure successful and reproducible experiments. First, careful selection of the appropriate antibody based on the specific application (WB, IHC, IP) and species reactivity is essential, as different antibodies show varying performance across applications . Second, thorough validation of antibody specificity using positive and negative controls, including NELFCD knockdown or overexpression systems, is necessary before proceeding with experimental work . Third, optimization of experimental conditions, including antibody dilution (typically 1:500-2000 for Western blot), incubation times, and detection methods, significantly impacts results . Fourth, proper storage at -20°C and avoidance of repeated freeze-thaw cycles preserves antibody activity . Finally, researchers should design experiments with appropriate normalization controls and statistical approaches to ensure valid interpretation of results. By addressing these considerations systematically, researchers can establish reliable protocols for investigating NELFCD's role in transcriptional regulation and disease processes.