The LINC00299 antibody is a specialized immunological reagent designed to detect the putative uncharacterized protein encoded by the long intergenic noncoding RNA 299 (LINC00299) gene. This antibody is primarily used in research to study LINC00299's role in cancer biology, immune regulation, and cellular processes .
Expression Analysis: LINC00299 expression is elevated in triple-negative breast cancer (TNBC) tissues (2.3-fold increase vs. adjacent nonmalignant tissue, p < 0.03) .
Biomarker Potential: Hypermethylation at cg06588802 in peripheral blood leukocytes (PBLs) correlates with TNBC risk in younger women (ages 22–46, p = 0.001) .
Clinical Associations: Higher LINC00299 expression in estrogen receptor-positive tumors (p < 0.05) .
Immune Regulation: LINC00299 interacts with the ID2 gene promoter, a key immune regulator, suggesting roles in immune cell modulation .
Atherosclerosis: The LINC00299/miR-490-3p axis regulates vascular smooth muscle cell proliferation and migration .
Gene | Sensitivity (%) | Specificity (%) | AUC | Cut-off |
---|---|---|---|---|
LINC00299 | 65 | 67 | 2.0 | 0.63 |
Data from breast cancer tissue analysis using RT-PCR .
LINC00299 is a long non-coding RNA that has gained attention in oncology research, particularly in breast cancer studies. It belongs to the growing class of functional RNAs that do not code for proteins but play regulatory roles in gene expression. Recent studies have demonstrated that LINC00299 expression is significantly upregulated (approximately 2.3-fold) in breast tumor tissues compared to adjacent non-malignant samples . This differential expression pattern suggests potential roles in cancer development and progression.
The significance of LINC00299 extends beyond expression differences, as it appears to be functionally linked to cellular stress response pathways. Research has revealed associations between LINC00299 and the unfolded protein response (UPR) pathway, specifically correlating with XBP1 splicing rates . Additionally, DNA methylation studies have identified LINC00299 hypermethylation as a potential biomarker for triple-negative breast cancer, particularly in younger women . These findings position LINC00299 as an important target for both basic research and potential clinical applications.
Researchers have access to several formats of LINC00299 antibodies, each optimized for specific experimental applications:
Biotin-conjugated polyclonal antibodies: These antibodies have biotin molecules attached to enhance detection sensitivity and compatibility with streptavidin-based detection systems. They are useful for experiments requiring signal amplification .
Non-conjugated polyclonal antibodies: These provide flexibility for researchers to use their preferred detection system, making them versatile for various applications including immunofluorescence studies .
Both types are primarily raised in rabbits using recombinant human putative uncharacterized protein encoded by LINC00299 (amino acids 2-105) as the immunogen, ensuring specificity for human samples . The polyclonal nature of these antibodies means they recognize multiple epitopes on the target protein, potentially increasing detection sensitivity while requiring careful validation to confirm specificity.
Commercial LINC00299 antibodies have specific characteristics that researchers should consider when designing experiments:
Characteristic | Specification | Notes |
---|---|---|
Antibody Type | Polyclonal | Recognizes multiple epitopes |
Host Species | Rabbit | Common host for research antibodies |
Species Reactivity | Human | Validated specifically for human samples |
Clonality | Polyclonal | Not derived from a single B-cell clone |
Isotype | IgG | Standard antibody class for research |
Purification Method | Protein G purification | >95% purity |
Applications | ELISA, IF | Validated for these specific techniques |
Recommended Dilution | IF: 1:50-1:200 | Application-specific dilution range |
Buffer Composition | 50% Glycerol, 0.01M PBS, pH 7.4, 0.03% Proclin 300 | Optimized for stability |
Form | Liquid | Ready-to-use format |
Storage Conditions | -20°C or -80°C | Avoid repeated freeze-thaw cycles |
This detailed information is derived from product documentation provided by antibody manufacturers . Understanding these specifications is crucial for selecting the appropriate antibody format and optimizing experimental conditions for specific research needs.
Proper handling and storage of LINC00299 antibodies are critical for maintaining their functionality and specificity:
Storage Recommendations:
Upon receipt, store antibodies at either -20°C or -80°C for long-term preservation .
Avoid repeated freeze-thaw cycles as this can lead to protein denaturation and loss of antibody function.
Consider aliquoting the antibody into single-use volumes before freezing to minimize freeze-thaw cycles.
The antibody is provided in a protective buffer containing 50% glycerol, 0.01M PBS (pH 7.4), and 0.03% Proclin 300 as a preservative, which helps maintain stability during storage .
Handling Guidelines:
Allow the antibody to thaw completely at 4°C before use.
Gently mix by inverting the tube several times (avoid vortexing to prevent protein denaturation).
Keep the antibody on ice during experimental setup.
Return unused antibody to -20°C or -80°C immediately after use.
When preparing working dilutions, use high-quality, sterile buffers appropriate for the intended application.
Following these handling and storage practices will help ensure consistent antibody performance and reproducible experimental results across multiple studies.
Optimizing immunofluorescence (IF) protocols for LINC00299 antibodies requires careful attention to several experimental parameters:
Dilution Optimization:
Start with the manufacturer's recommended dilution range (1:50-1:200) .
Perform a dilution series (e.g., 1:50, 1:100, 1:200) to determine the optimal antibody concentration for your specific samples.
Include appropriate controls at each dilution to assess background and specificity.
Protocol Optimization:
Fixation method: Test different fixation methods (e.g., 4% paraformaldehyde, methanol, or acetone) to determine which best preserves the epitope while maintaining tissue morphology.
Antigen retrieval: If working with FFPE tissues, evaluate different antigen retrieval methods (heat-induced versus enzymatic).
Blocking: Use 5-10% normal serum from the same species as the secondary antibody to reduce non-specific binding.
Incubation conditions: Compare overnight incubation at 4°C versus shorter incubations at room temperature.
Detection system: Select a fluorophore-conjugated secondary antibody appropriate for your microscopy setup.
Controls to Include:
Positive control (tissue known to express LINC00299)
Negative control (tissue known not to express LINC00299)
Secondary antibody-only control (omit primary antibody)
Isotype control (use non-specific rabbit IgG at the same concentration)
Systematic optimization with proper controls will help establish a reliable IF protocol for detecting LINC00299 in your specific experimental system.
Validating antibody specificity is crucial for generating reliable and reproducible research results. For LINC00299 antibodies, consider these validation approaches:
Genetic Approaches:
Knockdown/knockout validation: Compare staining between LINC00299 knockdown/knockout cells and wild-type controls. Specific antibodies should show reduced signal in knockdown/knockout systems.
Overexpression validation: Compare staining between cells overexpressing LINC00299 and control cells. Specific antibodies should show increased signal in overexpressing systems.
Biochemical Validation:
Western blot analysis: Verify that the antibody detects bands of the expected molecular weight.
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide before application to samples. Specific binding should be blocked by the immunizing peptide.
Mass spectrometry: Confirm the identity of immunoprecipitated proteins using mass spectrometry.
Cross-platform Validation:
Compare protein expression patterns detected by the antibody with mRNA expression data from RT-PCR or RNA-seq experiments.
Correlate antibody-based results with data from RNA in situ hybridization.
The combination of multiple validation approaches provides stronger evidence for antibody specificity than any single method alone, ensuring that experimental observations truly reflect LINC00299 biology rather than non-specific interactions.
Evidence from multiple studies positions LINC00299 as a significant factor in breast cancer biology:
Expression Analysis:
A cross-sectional study of Iranian breast cancer patients (2014-2019) demonstrated that LINC00299 expression was significantly upregulated (2.3-fold increase) in breast tumor tissues compared to adjacent non-malignant samples (p < 0.05) .
This expression pattern correlates with increased activity in cellular stress response pathways, suggesting functional relevance in cancer cell adaptation.
Pathway Integration:
LINC00299 expression shows association with the XBP1 splicing rate, a key marker of the unfolded protein response (UPR) pathway activation .
The UPR pathway is increasingly recognized as a critical mechanism for cancer cell survival under stress conditions, connecting LINC00299 to fundamental cancer cell biology.
Epigenetic Regulation:
DNA methylation studies have identified hypermethylation of the LINC00299 gene in triple-negative breast cancer (TNBC) .
Age-stratified analysis revealed that this hypermethylation pattern is more pronounced in younger women (age 26-52 at diagnosis and age 22-46 at blood draw) .
These findings collectively suggest that LINC00299 may participate in breast cancer development through epigenetic mechanisms and stress response pathways, with potential implications for understanding age-associated differences in disease presentation and progression.
LINC00299 methylation has emerged as a promising area in TNBC biomarker research:
Methodological Approaches:
Recent studies have employed MethyLight droplet digital PCR to quantify methylation at the cg06588802 site in LINC00299 .
This technique provides highly sensitive and quantitative assessment of DNA methylation patterns, enabling detection of subtle yet clinically relevant differences.
Key Findings:
Clinical Implications:
The age-dependent association suggests that LINC00299 hypermethylation may be particularly relevant as a biomarker in younger TNBC patients.
This finding aligns with observations that TNBC in younger women often exhibits distinct molecular features and more aggressive clinical behavior.
Blood-derived DNA methylation markers offer potential for non-invasive early detection and risk stratification.
These results highlight the importance of age-stratified analyses in biomarker research and suggest that LINC00299 methylation may contribute to the molecular landscape of early-onset TNBC.
The relationship between LINC00299 and the unfolded protein response (UPR) pathway represents an important mechanistic insight:
Experimental Evidence:
Studies measuring both LINC00299 expression and XBP1 splicing (XBP1s/u ratio) have identified a correlation between these molecular markers .
In breast cancer tissues, both LINC00299 expression (2.3-fold increase) and XBP1s/u ratio (2.8-fold increase) were significantly elevated compared to adjacent non-malignant tissues .
Mechanistic Context:
XBP1 splicing is a key event in the IRE1α branch of the UPR pathway, which is activated in response to endoplasmic reticulum stress.
Increased XBP1 splicing promotes survival under stress conditions by regulating genes involved in protein folding, quality control, and degradation.
The correlation between LINC00299 expression and XBP1s/u ratio suggests that this lncRNA may either regulate or respond to UPR activation.
Potential Regulatory Mechanisms:
LINC00299 may participate in regulating stress response pathways through various molecular mechanisms:
Direct interaction with XBP1 mRNA or protein
Modulation of IRE1α activity or expression
Regulation of genes involved in UPR signaling
Scaffold function for assembly of UPR-related protein complexes
Understanding this connection provides a framework for investigating how LINC00299 contributes to cancer cell adaptation to stress conditions, potentially identifying new therapeutic vulnerabilities in breast cancer.
Elucidating LINC00299's regulatory mechanisms requires sophisticated experimental approaches:
RNA-Protein Interaction Studies:
RNA Immunoprecipitation (RIP): Use antibodies against UPR pathway components (e.g., IRE1α, XBP1) to immunoprecipitate protein-RNA complexes, followed by qRT-PCR to detect LINC00299.
RNA Pull-down: Use biotinylated LINC00299 as bait to capture interacting proteins, followed by mass spectrometry identification.
CLIP-seq (Cross-linking immunoprecipitation-sequencing): Map RNA-protein interaction sites at single-nucleotide resolution to identify functional domains within LINC00299.
Loss-of-function and Gain-of-function Approaches:
Design CRISPR-Cas9 deletion constructs targeting different segments of LINC00299 to identify functional domains.
Generate LINC00299 overexpression models with mutations in predicted functional motifs.
Measure XBP1 splicing rates, UPR gene expression, and stress response dynamics following LINC00299 manipulation.
Subcellular Localization Studies:
RNA Fluorescence In Situ Hybridization (FISH): Determine LINC00299's subcellular distribution in normal versus cancer cells.
Subcellular fractionation: Quantify LINC00299 levels in nuclear, cytoplasmic, and endoplasmic reticulum fractions.
Co-localization analysis: Combine RNA FISH for LINC00299 with immunofluorescence for UPR components to assess spatial proximity.
Chromatin Association Studies:
ChIRP (Chromatin Isolation by RNA Purification): Map genomic binding sites of LINC00299.
ChIP-seq: Assess how LINC00299 manipulation affects chromatin accessibility and histone modifications at UPR-related genes.
These complementary approaches would provide comprehensive insights into whether LINC00299 functions primarily through chromatin modification, transcriptional regulation, post-transcriptional processing, or protein interaction mechanisms in breast cancer.
Translating LINC00299 research into clinical applications faces several significant challenges:
Biomarker Validation Challenges:
Standardization: Establishing standardized methods for measuring LINC00299 methylation or expression across different laboratories and platforms.
Cohort heterogeneity: The age-dependent associations observed in TNBC studies highlight the importance of carefully stratified analyses, requiring larger, diverse cohorts for validation.
Temporal dynamics: Determining whether LINC00299 alterations are early events in carcinogenesis or consequences of tumor progression.
Mechanistic Understanding Limitations:
Incomplete characterization of LINC00299's molecular interactions and downstream effects.
Uncertainty about tissue-specific functions and context-dependent regulation.
Limited understanding of how LINC00299 interactions with the UPR pathway might differ between normal and malignant cells.
Therapeutic Development Barriers:
Technical challenges in targeted modulation of lncRNAs in vivo.
Potential off-target effects due to sequence similarities with other lncRNAs.
Development of appropriate delivery systems that can reach target tissues.
Identification of patient subgroups most likely to benefit from LINC00299-targeted approaches.
Research Infrastructure Needs:
Integration of LINC00299 data into comprehensive lncRNA databases like LncRNADisease .
Development of computational tools to predict LINC00299 functions and interactions in different cellular contexts.
Establishment of appropriate preclinical models that recapitulate LINC00299's roles in human disease.
Addressing these challenges requires coordinated efforts across basic, translational, and clinical research domains to fully realize the potential of LINC00299 as a biomarker or therapeutic target.
Integrating LINC00299 research into broader lncRNA regulatory networks requires multilevel data integration approaches:
Database Integration:
Leverage specialized lncRNA databases like LncRNADisease to place LINC00299 findings in context with other lncRNA-disease associations.
Cross-reference LINC00299 interactions with data from platforms documenting RNA-protein, RNA-DNA, and RNA-RNA interactions.
Utilize GWAS databases to identify potential genetic variants affecting LINC00299 function or expression .
Network Analysis Methods:
Co-expression network analysis: Identify lncRNAs and mRNAs whose expression patterns correlate with LINC00299 across different conditions.
Pathway enrichment analysis: Determine biological processes and signaling pathways associated with LINC00299 and its co-expressed genes.
Protein-lncRNA interaction networks: Map how LINC00299 fits into known protein interaction networks, particularly in UPR pathway regulation.
Multi-omics Integration:
Combine transcriptomic (RNA-seq), epigenomic (DNA methylation, histone modifications), and proteomic data to build comprehensive regulatory models.
Analyze how LINC00299 alterations correlate with changes in other molecular layers.
Develop computational methods to predict functional consequences of LINC00299 dysregulation across different cellular contexts.
Comparative Analysis Across Diseases:
Compare LINC00299's role in breast cancer with its potential functions in other cancer types or diseases.
Identify common and tissue-specific mechanisms of LINC00299 regulation and function.
Assess whether findings in triple-negative breast cancer translate to other aggressive cancer subtypes.
This integrative approach would place LINC00299 research within a systems biology framework, enhancing our understanding of how this lncRNA contributes to complex regulatory networks in health and disease.
Selecting appropriate detection methods for LINC00299 research requires understanding the strengths and limitations of each technique:
Method | Advantages | Limitations | Best Applications |
---|---|---|---|
RT-PCR | High sensitivity, quantitative, relatively low cost, widely accessible | Cannot provide spatial information, susceptible to contamination | Expression quantification, screening studies |
RNA-seq | Comprehensive, captures isoforms, allows novel discovery, unbiased | Higher cost, complex analysis, lower sensitivity for low-abundance transcripts | Transcriptome-wide analysis, isoform discovery |
RNA FISH | Provides spatial information, single-cell resolution, detects subcellular localization | Labor-intensive, requires specialized equipment, lower throughput | Subcellular localization studies, heterogeneity analysis |
Methylation Analysis (e.g., MethyLight ddPCR) | Highly sensitive for epigenetic changes, works with cell-free DNA | Provides no information on expression or function | Biomarker development, early detection studies |
Antibody-based Methods (IF, IHC) | Protein-level detection, spatial information, compatible with FFPE samples | Dependent on antibody specificity, indirect measure of RNA | Protein expression studies, clinical specimens |
For comprehensive characterization of LINC00299, researchers should consider combining multiple approaches. For example, RT-PCR can be used for initial expression screening across samples, followed by RNA FISH for detailed subcellular localization in cells of interest, and methylation analysis for evaluating potential biomarker applications .
Robust experimental design with appropriate controls is essential for generating reliable data on LINC00299:
Cell Line Studies:
Positive controls: Include cell lines with known high LINC00299 expression (e.g., certain breast cancer cell lines based on previous studies).
Negative controls: Include cell lines with minimal LINC00299 expression.
Knockdown validation: Generate LINC00299 knockdown lines to serve as biological negative controls.
Overexpression controls: Generate stable LINC00299 overexpression lines for functional studies.
Tissue Sample Studies:
Technical controls: Include reference gene assays (e.g., GAPDH, β-actin) to normalize for RNA quality and quantity.
Matched controls: Whenever possible, use adjacent non-malignant tissue from the same patient as internal controls.
Age-matched controls: Given the age-dependent associations observed in previous studies , ensure proper age stratification in case-control designs.
Methylation Studies:
Unmethylated controls: Include samples known to have low methylation at the target sites.
Fully methylated controls: Use in vitro methylated DNA as positive controls.
Non-converted DNA controls: Include controls to assess bisulfite conversion efficiency.
Specificity Controls for Antibody-based Studies:
Peptide competition: Pre-incubate antibodies with immunizing peptides to demonstrate specificity.
Isotype controls: Use non-specific IgG from the same species as the primary antibody.
Secondary-only controls: Omit primary antibody to assess background from secondary antibody.
Implementing these comprehensive control strategies will enhance data reliability and facilitate comparison of results across different experimental systems and research groups.