The NME1 Antibody Pair is a specialized immunological reagent designed for the detection and quantification of the NME1 protein (also known as NM23A or non-metastatic cells 1, protein). This paired antibody system consists of two antibodies—one for capturing the target antigen (capture antibody) and another for detecting it (detection antibody). The pair is commonly used in sandwich enzyme-linked immunosorbent assays (ELISA), immunoprecipitation, and other immunoassays.
Capture Antibody: Typically a polyclonal or recombinant antibody that binds to a specific epitope of NME1.
Detection Antibody: Often a monoclonal antibody conjugated to a reporter enzyme (e.g., HRP) or fluorescent tag for signal amplification.
Applications: Quantification of NME1 in biological samples (e.g., serum, lysates) and research into metastasis suppression mechanisms .
Parameter | Details |
---|---|
Host/Isotype | Rabbit IgG (capture), Rabbit IgG (detection) |
Reactivity | Human |
Immunogen | NME1 fusion protein (Ag1548) |
Applications | Sandwich ELISA, multiplex bead-based assays, Simoa® immunoassays |
Range (ELISA) | 3.13–200 ng/mL |
Conjugation | Unconjugated (ready for custom labeling) |
Purification | Protein A purification |
Storage Buffer | PBS only (no preservatives) |
Catalog Number | MP01750-3 (Proteintech) |
Parameter | Details |
---|---|
Host/Isotype | Rabbit MaxPab® (capture), Mouse Monoclonal IgG1 kappa (detection) |
Reactivity | Human |
Antigen | NME1 |
Applications | ELISA, immunoprecipitation, Western blot |
Catalog Number | 10540-636EA (VWR) |
Supplier | Abnova |
NME1 is a well-characterized metastasis suppressor gene whose expression correlates with reduced metastatic potential in cancers (e.g., melanoma, breast carcinoma) . The antibody pair has been validated in studies examining:
Gene Expression Profiling: NME1 regulates metastasis-related genes (e.g., MMP1, ANGPT2) .
Protein-Protein Interactions: NME1 interacts with dynamin 2 (DNM2) to modulate endocytosis and tumor cell motility .
ELISA Sensitivity: The Proteintech pair achieves a detection limit of 3.13 ng/mL for NME1 in serum or lysates .
Cross-Reactivity: Minimal cross-reactivity with non-target proteins (validated via Western blot) .
Dynamin 2 (DNM2) Modulation: NME1 enhances DNM2-mediated endocytosis, reducing tumor cell surface receptor signaling (e.g., EGFR) .
Prognostic Biomarker Potential: High NME1 expression correlates with improved survival in breast cancer subtypes .
NME1 functions primarily as a metastasis suppressor gene that significantly reduces metastasis without affecting primary tumor growth. It exhibits reduced expression in metastatic cancers and has the ability to inhibit metastatic properties of melanoma and other cancer cells . NME1 exerts its metastasis suppression through several mechanisms:
Direct activation of transcription by binding to promoter regions of target genes like ALDOC
Promotion of dynamin 2 (DNM2) oligomerization and GTPase activity, affecting endocytosis of receptors such as transferrin and EGF receptors
Inhibition of epithelial-mesenchymal transition (EMT), a critical early step in metastasis
Regulation of cellular motility and invasion through multiple molecular pathways
The nucleoside diphosphate kinase activity of NME1 appears important for some functions, but studies have shown that certain metastasis-suppressive activities, such as promoting endocytosis, require additional enzymatic activities like histidine protein kinase function .
NME1 Antibody Pairs typically consist of two matched antibodies designed for specific and quantitative detection of human NME1 protein . The antibody pair system involves:
A capture antibody: Usually rabbit polyclonal antibodies that are affinity-purified (e.g., Rabbit MaxPab® affinity purified Polyclonal Anti-NME1)
A detection antibody: Typically mouse monoclonal antibodies (e.g., Mouse Monoclonal Anti-NME1, IgG1 kappa)
This dual-antibody approach enables highly specific detection of NME1 in various experimental settings, including:
Quantitative assessment of NME1 protein levels in cancer cell lines
Evaluation of metastatic potential based on NME1 expression
Investigation of NME1's interactions with other proteins (e.g., DNM2)
Monitoring changes in NME1 expression after experimental manipulations
Correlation of NME1 levels with other markers of cancer progression
The specificity of the antibody pair allows researchers to reliably detect and quantify NME1 across different experimental conditions.
Validation of NME1 antibodies is crucial for experimental reliability. Recommended validation approaches include:
Positive and negative control samples: Use cell lines with known high NME1 expression (e.g., non-metastatic cell lines) versus those with low expression (e.g., metastatic cell lines)
Genetic validation: Compare antibody detection in wildtype cells versus NME1-knockout cells generated via CRISPR-Cas9 gene editing
Antibody specificity: Confirm that anti-NME1 antibodies do not cross-react with the highly homologous NME2 protein by testing in NME2-knockout models
Protein knockdown validation: Compare detection in control cells versus cells treated with siRNA against NME1
Multiple detection methods: Verify results using different antibody-based techniques (western blot, immunofluorescence, flow cytometry)
In studies where both NME1 and NME2 are being investigated, it's critical to ensure that selective loss of NME1 or NME2 does not alter the protein level of the other isoform, as their expression appears to be regulated independently .
Robust experimental designs to study NME1's metastasis suppression functions include:
Gene manipulation strategies:
Functional assays:
Cell motility and migration assays to assess NME1's impact on cellular movement
Endocytosis assays tracking internalization of transferrin and EGF receptors
EMT marker analysis via western blotting and flow cytometry to measure epithelial markers (E-cadherin, cytokeratin 18, β-catenin) and mesenchymal markers (N-cadherin, vimentin)
Aggregation and dispersion assays to evaluate E-cadherin-mediated cell-cell adhesion
In vivo models:
These approaches should be combined for comprehensive assessment of NME1's metastasis suppression mechanisms in specific cancer contexts.
Despite their high sequence homology, NME1 and NME2 have distinct functions in cancer progression. To differentiate their roles:
Generate separate genetic models:
Conduct parallel phenotypic analysis:
Evaluate isoform-specific molecular functions:
Research has demonstrated that NME1, but not NME2, functions as a powerful inhibitor of EMT , highlighting the importance of studying these isoforms independently.
To effectively study NME1's direct role in transcriptional regulation:
Promoter activity assessment:
Chromatin immunoprecipitation (ChIP):
Transcription analysis:
Mechanistic validation:
These approaches collectively provide robust evidence for direct transcriptional regulatory functions of NME1.
Interpreting EMT marker changes requires comprehensive analysis of multiple markers and phenotypic characteristics:
Expression pattern analysis:
Marker interpretation table:
Functional validation:
Contextual considerations:
Different cancer types may show varying dependency on specific EMT markers
The hybrid EMT state may be more relevant for metastasis than complete EMT
To effectively study NME1's role in endocytosis and signaling:
Receptor internalization assays:
Endocytic pathway analysis:
Signaling pathway investigation:
Mechanistic studies:
These approaches provide mechanistic insights into how NME1 modulates receptor trafficking and signaling through dynamin-dependent pathways.
Variations in NME1 expression and function across cancer types require careful interpretation:
Context-dependent analysis:
Multi-level validation:
Confirm RNA expression changes with protein-level analysis
Correlate expression data with functional metastasis assays
Validate clinical relevance with patient outcome data
Methodological consistency:
Standardize detection methods across studies
Use both relative and absolute quantification where possible
Employ multiple antibodies or detection approaches to confirm findings
Consider regulatory mechanisms:
Evaluate post-translational modifications of NME1 that may affect function without changing expression levels
Assess subcellular localization of NME1, as nuclear versus cytoplasmic localization may have different implications
Analyze NME1 in relation to key partners:
Advanced applications of NME1 antibodies in treatment response studies include:
Predictive biomarker development:
Screen patient tumor samples pre-treatment to correlate NME1 levels with response outcomes
Develop quantitative immunohistochemistry protocols using antibody pairs for clinical applications
Create standardized scoring systems for NME1 expression in tissue samples
Treatment-induced changes:
Monitor NME1 expression dynamics during treatment course
Assess whether therapy-resistant populations show altered NME1 expression
Investigate if combination therapies affect NME1 levels or localization
Mechanistic studies:
Therapeutic targeting approaches:
Use antibody pairs to screen compounds that may restore NME1 expression in metastatic cells
Develop assays to identify molecules that enhance NME1's metastasis-suppressive functions
Validate effects of putative NME1-targeting therapies on downstream pathways
NME1's impact on epigenetic regulation requires specialized experimental approaches:
Chromatin modification analysis:
Mechanistic investigation:
Determine if NME1 directly interacts with chromatin modifiers or remodeling complexes
Analyze recruitment of epigenetic enzymes to NME1-bound genomic regions
Assess whether NME1's enzymatic activities are required for epigenetic effects
Target gene validation:
Functional consequences:
Link epigenetic alterations to relevant cancer phenotypes (migration, invasion, EMT)
Determine if pharmacological modulation of the epigenetic landscape can mimic or reverse NME1's effects
Investigate whether NME1's epigenetic functions are tissue- or context-specific
Translating NME1 research from 2D cultures to more physiologically relevant models requires:
3D culture systems:
In vivo metastasis models:
Develop orthotopic implantation models with NME1-manipulated cells
Track metastatic spread using imaging technologies
Compare lung colonization efficiency between control and NME1-manipulated cells
Validate the dependency of metastasis suppression on key mechanisms (e.g., using DNM2 knockdown cells)
Genetically engineered mouse models:
Translational considerations:
Develop methods to detect and quantify NME1 in patient-derived xenografts
Analyze circulating tumor cells for NME1 expression and EMT status
Correlate NME1 expression patterns with metastatic burden in animal models