NME1 Antibody Pair

Shipped with Ice Packs
In Stock

Description

Introduction to NME1 Antibody Pair

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.

Key Features:

  • 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 .

2.1. Proteintech Recombinant Matched Antibody Pair (MP01750-3)

ParameterDetails
Host/IsotypeRabbit IgG (capture), Rabbit IgG (detection)
ReactivityHuman
ImmunogenNME1 fusion protein (Ag1548)
ApplicationsSandwich ELISA, multiplex bead-based assays, Simoa® immunoassays
Range (ELISA)3.13–200 ng/mL
ConjugationUnconjugated (ready for custom labeling)
PurificationProtein A purification
Storage BufferPBS only (no preservatives)
Catalog NumberMP01750-3 (Proteintech)

2.2. Abnova Antibody Pair Set (H00004830-AP11)

ParameterDetails
Host/IsotypeRabbit MaxPab® (capture), Mouse Monoclonal IgG1 kappa (detection)
ReactivityHuman
AntigenNME1
ApplicationsELISA, immunoprecipitation, Western blot
Catalog Number10540-636EA (VWR)
SupplierAbnova

3.1. Metastasis Suppression Studies

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 .

3.2. Assay Performance

  • 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) .

4.1. Metastasis Suppression Mechanisms

  • 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 .

4.2. Antibody Pair Performance in Assays

  • Sandwich ELISA: Achieves a linear range of 3.13–200 ng/mL with <10% intra-assay CV .

  • Immunoprecipitation: Efficiently pulls down NME1-DNM2 complexes for Western blot analysis .

Product Specs

Buffer
**Capture:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders 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 timelines.
Notes
We recommend using the capture antibody at a concentration of 0.5 µg/mL and the detection antibody at a concentration of 0.25 µg/mL. Optimal dilutions should be determined experimentally by the researcher.
Synonyms
NDK A,NDP kinase A,A-activated DNase,GAAD,Metastasis inhibition factor nm23,Tumor metastatic process-associated protein,nm23-H1
Target Names
NME1

Q&A

What is NME1 and what are its primary biological functions in cancer?

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 .

How do NME1 Antibody Pairs work and what are their typical research applications?

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.

How can researchers validate the specificity of NME1 antibodies in their experimental systems?

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 .

What experimental approaches are most effective for studying NME1's role in metastasis suppression?

Robust experimental designs to study NME1's metastasis suppression functions include:

  • Gene manipulation strategies:

    • Overexpression of NME1 in metastatic cell lines that have lost NME1 expression

    • Silencing of NME1 in non-metastatic cell lines with normal NME1 expression

    • CRISPR-Cas9 gene editing for complete inactivation of NME1

  • 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:

    • Lung metastasis assays comparing NME1-overexpressing cells with control cells

    • Analysis of NME1 knockout in mouse models of ultraviolet light-induced melanoma

These approaches should be combined for comprehensive assessment of NME1's metastasis suppression mechanisms in specific cancer contexts.

How should researchers design experiments to differentiate between NME1 and NME2 functions?

Despite their high sequence homology, NME1 and NME2 have distinct functions in cancer progression. To differentiate their roles:

  • Generate separate genetic models:

    • Create cell lines with selective knockout of either NME1 or NME2 using CRISPR-Cas9 gene editing with isoform-specific guide RNAs

    • Confirm absence of compensatory expression changes (validate that loss of one isoform doesn't affect expression of the other)

  • Conduct parallel phenotypic analysis:

    • Compare EMT marker expression between NME1-ablated, NME2-ablated, and control cells

    • Analyze cellular morphology changes across all three conditions

    • Assess cell-surface expression of key proteins like E-cadherin using flow cytometry

  • Evaluate isoform-specific molecular functions:

    • Test the effects of both NME1 and NME2 on dynamin 2 oligomerization and GTPase activity

    • Analyze the impact of each isoform on endocytosis of receptors

    • Measure effects on signaling pathways like AKT and MAPK pathways

Research has demonstrated that NME1, but not NME2, functions as a powerful inhibitor of EMT , highlighting the importance of studying these isoforms independently.

What methodological considerations are important when investigating NME1's transcriptional regulatory functions?

To effectively study NME1's direct role in transcriptional regulation:

  • Promoter activity assessment:

    • Generate promoter-luciferase constructs for putative NME1 target genes (e.g., ALDOC)

    • Compare promoter activity in NME1-overexpressing versus control cells

  • Chromatin immunoprecipitation (ChIP):

    • Use validated anti-NME1 antibodies for immunoprecipitation of protein-DNA complexes

    • Analyze NME1 occupancy at target gene promoters (both upstream and proximal regions)

    • Include parallel investigation of epigenetic activation markers (H3K4me3 and H3K27ac)

    • Assess RNA polymerase II recruitment

  • Transcription analysis:

    • Measure both pre-mRNA and mature mRNA levels to distinguish direct transcriptional effects from post-transcriptional regulation

    • Compare expression at the protein level via western blotting

  • Mechanistic validation:

    • Use site-directed mutagenesis to create NME1 variants lacking specific enzymatic functions (e.g., histidine protein kinase activity versus nucleoside diphosphate kinase activity)

    • Test these variants in functional assays to determine which activities are required for transcriptional regulation

These approaches collectively provide robust evidence for direct transcriptional regulatory functions of NME1.

How should researchers interpret changes in EMT marker expression after NME1 manipulation?

Interpreting EMT marker changes requires comprehensive analysis of multiple markers and phenotypic characteristics:

  • Expression pattern analysis:

    • Complete EMT is characterized by loss of all epithelial markers and gain of all mesenchymal markers

    • Hybrid or partial EMT (often associated with high metastatic potential) shows retention of some epithelial markers alongside acquisition of mesenchymal traits

  • Marker interpretation table:

Marker TypeMarkersIn NME1-ablated CellsIn NME1-overexpressing CellsInterpretation
EpithelialE-cadherinDecreased (protein level and cell surface localization) IncreasedNME1 promotes epithelial phenotype
EpithelialCytokeratin 18, β-cateninNo change No significant changeIncomplete EMT upon NME1 loss
MesenchymalN-cadherin, VimentinIncreased DecreasedNME1 suppresses mesenchymal transition
EMT-TFsZEB1Markedly increased DecreasedNME1 regulates key EMT transcription factors
EMT-TFsSlug (SNAI2)Variable increase DecreasedClone-dependent regulation by NME1
  • Functional validation:

    • Correlate marker changes with functional assessments (e.g., cell-cell adhesion, motility)

    • Consider that NME1 loss often leads to a "hybrid phenotype" between epithelial and mesenchymal states, which is associated with high metastatic potential

  • 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

What approaches are recommended for studying NME1's effect on receptor endocytosis and signaling?

To effectively study NME1's role in endocytosis and signaling:

  • Receptor internalization assays:

    • Track fluorescently-labeled transferrin or EGF uptake in cells with manipulated NME1 levels

    • Quantify co-localization with endocytic markers like Rab5

    • Measure depletion of adapter proteins (e.g., AP2) from the cell surface

  • Endocytic pathway analysis:

    • Assess Rab5-GTP levels as indicators of endocytosis progression

    • Evaluate the impact of dynamin inhibitors (e.g., Iminodyn-22, Dynole-34-2) on NME1-induced endocytosis

    • Use shRNA-mediated knockdown of dynamin 2 to determine its necessity for NME1's effects

  • Signaling pathway investigation:

    • Use the EGF-EGFR signaling axis as a model system

    • Measure phosphorylation levels of EGFR and downstream effectors like Akt

    • Compare signaling dynamics in control, NME1-overexpressing, and NME1-overexpressing/DNM2-knockdown conditions

  • Mechanistic studies:

    • Perform co-immunoprecipitation to confirm NME1-DNM2 interaction

    • Assess DNM2 oligomerization in the presence/absence of NME1

    • Measure DNM2 GTPase activity with purified proteins

These approaches provide mechanistic insights into how NME1 modulates receptor trafficking and signaling through dynamin-dependent pathways.

How can researchers account for potential contradictions in NME1 expression data across different cancer types?

Variations in NME1 expression and function across cancer types require careful interpretation:

  • Context-dependent analysis:

    • Compare NME1 expression within the same cancer type at different progression stages

    • Consider tissue-specific microenvironmental factors that may influence NME1 function

    • Account for molecular subtypes (e.g., triple-negative versus ER-positive breast cancers)

  • 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:

    • Measure expression of interaction partners like dynamin 2

    • Evaluate downstream effectors that may be differentially regulated in various cancer contexts

How can NME1 Antibody Pairs be utilized to investigate the relationship between NME1 and cancer treatment response?

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:

    • Determine if treatments affect NME1's interaction with key partners like dynamin 2

    • Analyze how treatments impact NME1-regulated pathways such as AKT and MAPK signaling

    • Evaluate if NME1 status affects drug-induced changes in EMT markers

  • 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

What considerations are important when designing experiments to study the epigenetic effects of NME1?

NME1's impact on epigenetic regulation requires specialized experimental approaches:

  • Chromatin modification analysis:

    • Perform ChIP-seq for histone modifications (H3K4me3, H3K27ac) in the presence/absence of NME1

    • Compare broad epigenetic landscapes using techniques like ATAC-seq to assess chromatin accessibility

    • Investigate DNA methylation patterns at NME1-regulated promoters

  • 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:

    • Perform genome-wide analyses to identify all promoters exhibiting NME1-dependent epigenetic changes

    • Validate findings at individual loci using focused ChIP-qPCR for both NME1 and epigenetic marks

    • Correlate epigenetic changes with transcriptional outcomes

  • 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

How should researchers approach the study of NME1 in three-dimensional and in vivo models?

Translating NME1 research from 2D cultures to more physiologically relevant models requires:

  • 3D culture systems:

    • Establish spheroid or organoid models with manipulated NME1 expression

    • Assess epithelial organization, polarity, and invasive capacity

    • Compare NME1's effects on EMT markers in 2D versus 3D contexts

  • 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:

    • Utilize tissue-specific Nme1 knockout approaches

    • Combine with relevant oncogenic drivers for specific cancer types

    • Consider the role of Nme1-Nme2 locus in UV-induced melanoma 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

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.